Language selection

Search

Patent 2877397 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2877397
(54) English Title: ANTIBODIES TO TAU
(54) French Title: ANTICORPS DIRIGES CONTRE TAU
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 16/18 (2006.01)
  • G01N 33/53 (2006.01)
(72) Inventors :
  • HOLTZMAN, DAVID (United States of America)
  • JIANG, HONG (United States of America)
  • DIAMOND, MARC (United States of America)
  • KFOURY, NAJLA (United States of America)
  • HOLMES, BRANDON (United States of America)
(73) Owners :
  • WASHINGTON UNIVERSITY (United States of America)
(71) Applicants :
  • WASHINGTON UNIVERSITY (United States of America)
(74) Agent: TORYS LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2013-07-03
(87) Open to Public Inspection: 2014-01-09
Examination requested: 2018-06-28
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2013/049333
(87) International Publication Number: WO2014/008404
(85) National Entry: 2014-12-18

(30) Application Priority Data:
Application No. Country/Territory Date
61/667,515 United States of America 2012-07-03
61/694,989 United States of America 2012-08-30

Abstracts

English Abstract

This invention relates to antibodies to tau and methods of use thereof.


French Abstract

Cette invention concerne des anticorps dirigés contre tau et leurs procédés d'utilisation.

Claims

Note: Claims are shown in the official language in which they were submitted.


IN THE CLAIMS
What is claimed is:
1. An isolated antibody, wherein the antibody specifically binds tau and
recognizes
an epitope within an amino acid sequence selected from the group consisting of
SEQ ID
NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6,
SEQ ID NO: 7, and SEQ ID NO: 8.
2. The isolated antibody of claim 1, wherein the antibody comprises an
amino acid
sequence selected from the group consisting of SEQ ID NO: 14 and SEQ ID NO:
15.
3. The isolated antibody of claim 1, wherein the antibody is encoded by a
nucleic
acid sequence comprising a nucleic acid sequence selected from the group
consisting
of SEQ ID NO: 12 and SEQ ID NO: 13.
4. An isolated antibody, wherein the antibody specifically binds tau and
comprises a
light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 18 with zero
to
two amino acid substitutions.
5. An isolated antibody, wherein the antibody specifically binds tau and
comprises a
heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 21 with zero
to
two amino acid substitutions.
6. An isolated antibody of any of the preceding claims, wherein the
antibody is
selected from the group consisting of a single-chain antibody, an antibody
fragment, a
chimeric antibody, or a humanized antibody.
7. An isolated antibody of any of the preceding claims wherein the antibody
is
specifically able to block tau seeding activity in a cellular tau aggregation
assay.
102

8. A method for reducing the spread of tau aggregation in the brain of a
subject,
the method comprising administering a pharmacologically effective amount of
anti-tau
antibody to the subject, wherein the anti-tau antibody is an isolated antibody
of any of
the preceding claims.
9. The method of claim 8, wherein the method further comprises improving in
a
subject at least one symptom associated with tau aggregation.
10. The method of claim 9, wherein the at least one symptom associated with
tau
aggregation is selected from the group consisting of tau pathology, impaired
cognitive
function, altered behavior, abnormal language function, emotional
dysregulation,
seizures, impaired nervous system structure or function, and an increased risk
of
development of Alzheimer's disease.
11. The method of claim 8, wherein the administration comprises an
effective
systemic route of administration.
12. The method of claim 8, wherein the administration comprises an
effective local
route of administration, including directly within the central nervous system.
13. An immunoassay comprising at least two isolated antibodies of any of
the
preceding claims 1-7.
14. The immunoassay of claim 13, wherein the immunoassay comprises at least
two
captures antibodies and a detection antibody, and wherein each capture
antibody is an
isolated anti-tau antibody that recognizes a tau epitope distinct from the
other.
15. The immunoassay of claim 14, wherein a first capture antibody is an
isolated
antibody that specifically binds tau and recognizes an epitope within SEQ ID
NO: 7, a
second capture antibody is an isolated antibody that specifically binds tau
and
103

recognizes an epitope within SEQ ID NO: 8, and a detection antibody is an
isolated
antibody that specifically binds tau and recognizes an epitope within SEQ ID
NO: 8.
16. A method of measuring the amount of tau aggregate in a sample of a
biological
fluid obtained from a subject, the method comprising measuring the amount of
tau
aggregate using an immunoassay of any of claims 13-15.
17. The method of claim 16, wherein the tau aggregate is immunoprecipitated
from
the sample using an isolated anti-tau antibody and then the amount of
immunoprecipitated tau aggregate is measured.
104

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
ANTIBODIES TO TAU
GOVERNMENT SUPPORT
[0001] This invention was made with government support under
1R01NS071835 awarded by National Institute of Neurological Disorders and
Stroke.
The government has certain rights in the invention.
FIELD OF THE INVENTION
[0002] This invention relates to antibodies to tau and methods of
use
thereof.
BACKGROUND OF THE INVENTION
[0003] Aggregation of the microtubule associated protein tau is
associated
with several neurodegenerative disorders, including Alzheimer's disease (AD)
and
frontotemporal dementia. In AD, pathological tau aggregation spreads
progressively
throughout the brain, possibly along existing neural networks. AD is the most
common
cause of dementia and is an increasing public health problem. It is currently
estimated
to afflict 5 million people in the United States, with an expected increase to
13 million by
the year 2050. Alzheimer's Disease leads to loss of memory, cognitive
function, and
ultimately loss of independence. It takes a heavy personal and financial toll
on the
patient and the family. Because of the severity and increasing prevalence of
the AD and
other neurodegenerative diseases associated with aggregation of tau in the
population,
it is urgent that better treatments and detection methods be developed.
REFERENCE TO COLOR FIGURES
[0004] The application file contains at least one photograph
executed in
color. Copies of this patent application publication with color photographs
will be
provided by the Office upon request and payment of the necessary fee.
1

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
BRIEF DESCRIPTION OF THE FIGURES
[0005] FIG. 1 depicts the amino acid sequence for N-terminal (A)
and C-
terminal (B) human tau (htau).
[0006] FIG. 2 depicts graphs showing the KD for HJ8.1 towards Human
Tau (A) and Mouse Tau (B).
[0007] FIG. 3 depicts graphs showing the KD for HJ8.2 towards Human
Tau (A) and Mouse Tau (B).
[0008] FIG. 4 depicts graphs showing the KD for HJ8.3 towards Human
Tau (A) and Mouse Tau (B).
[0009] FIG. 5 depicts graphs showing the KD for HJ8.4 towards Human
Tau (A) and Mouse Tau (B).
[0010] FIG. 6 depicts graphs showing the KD for HJ8.5 towards Human
Tau (A) and Mouse Tau (B).
[0011] FIG. 7 depicts graphs showing the KD for HJ8.7 towards Human
Tau (A) and Mouse Tau (B).
[0012] FIG. 8 depicts graphs showing the KD for HJ8.8 towards Human
Tau (A) and Mouse Tau (B).
[0013] FIG. 9 depicts graphs showing the KD for HJ9.1 towards Human
Tau (A) and Mouse Tau (B).
[0014] FIG. 10 depicts graphs showing the KD for HJ9.2 towards
Human
Tau (A) and Mouse Tau (B).
[0015] FIG. 11 depicts graphs showing the KD for HJ9.3 towards
Human
Tau (A) and Mouse Tau (B).
[0016] FIG. 12 depicts graphs showing the KD for HJ9.4 towards
Human
Tau (A) and Mouse Tau (B).
[0017] FIG. 13 depicts graphs showing the KD for HJ9.5 towards
Human
Tau (A) and Mouse Tau (B).
[0018] FIG. 14 depicts immunoblots showing the presence of full-
length
tau in ISF of wild-type and P301S tg mice. (A) Hippocampal lysates from Tau KO
(KO),
wild-type (WT), and P301S tg (P301S tg) mice were analyzed by immunoblot with
the
2

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
anti-tau antibody BT-2 or anti-actin antibody. Thirteen micrograms of protein
were
loaded per well. Four bands corresponding to endogenous murine tau and one
band
corresponding to human tau are indicated as white circles and a black circle,
respectively. There is also a 39 kDa band representing a form of human tau in
the
P301S tg hippocampal lysate. This may represent a tau degradation product. ISF
tau
from wild-type (WT) and P3015 tg (P301S tg) mice was immunoprecipitated by
anti-tau
monoclonal antibodies HJ9.3 (B) or HJ8.1 (C) and analyzed by immunoblot. The
bands
were visualized by biotinylated BT-2 antibody. The gray and black arrows
indicate
endogenous murine tau and human tau, respectively.
[0019] FIG. 15 (A) illustrates a schematic representation of the
different
mutant tau constructs used in this study, and (B-D) depict images showing Tau
RD
proteins form fibrillar aggregates in transfected HEK293 cells. (A) Depending
on the
experimental design, each form of mutant tau was either fused at the carboxyl
terminus
to cyan or yellow fluorescent protein (CFP or YFP), or to a hemagglutinin (HA)
tag. (B)
Atomic force microscopy (AFM) performed on SDS-insoluble material from HEK293
cells transiently transfected with the various forms of RD reveals that RD(AK)-
HA and
RD(LM)-HA produced obvious fibrillar species. No fibrils were detected in the
aggregation-resistant RD(PP)-HA. (n=2), Scale bars, lpm. (C) HEK293 cells
transiently
transfected with the various forms of RD-YFP and YFP alone were stained with X-
34,
an amyloid-specific dye. Inclusions formed by RD(wt)-YFP, RD(AK)-YFP and
RD(LM)-
YFP, visualized by confocal microscopy, also stained positive for X-34. No X-
34 positive
cells were detected upon expression of YFP alone or RD(PP)- YFP. Arrows
indicate
inclusions stained with X-34. (n=3) (D) Non-Transfected cells (NT) and various
forms of
RD-YFP/CFP were transfected into HEK293 cells, followed by Triton/SDS
extraction
and Western blotting using an antibody against the RD region. Both monomer and

higher order molecular weight species were detected. (S. Soluble protein and
P. Pellet
insoluble protein). This was repeated three times with identical results.
[0020] FIG. 16 shows Tau RD aggregates in HEK293 cells are detected
by
FRET. To quantitate intracellular RD protein aggregation by fluorescence
resonance
energy transfer (FRET), various RD mutants (wt, AK, PP, LM) fused to YFP and
CFP
3

CA 02877397 2014-12-18
WO 2014/008404
PCT/US2013/049333
were co-transfected into HEK293 cells. (A). HEK293 cells co-transfected with
RD(LM)-
CFP/YFP were imaged and intracellular aggregate formation was quantified using

FRET acceptor photobleaching microscopy. Donor signal before (Pre) and after
(Post)
acceptor photobleaching confirmed that RD(LM)-CFP/ YFP inclusions produced a
mean
FRET efficiency of 18.2% 0.058 SD (n=6). The upper and lower panels depict
the
acceptor and donor channels, respectively, before and after photobleaching.
The top
right image is a representative heat map of the calculated FRET efficiency.
The scale
bar of the histogram depicts the calculated FRET efficiency on a pixel-by-
pixel basis.
The FRET efficiency of Tau RD aggregate was -34% in this cell. (B). Using a
FPR,
relative FRET from various constructs was determined. No significant FRET from

RD(PP)-CFP/YFP was observed. However, RD(AK)-CFP/YFP and RD(LM)-CFP/YFP
each produced a strong FRET signal (n=3). (C). HEK293 cells expressing RD(AK)-
CFP/YFP were exposed to various concentrations of RD(wt)-HA fibrils (monomer
equivalents of 0.01, 0.03, 0.1 and 0.3 pM) for 9 h. Extracellular RD(wt)-HA
fibrils dose-
dependently induced aggregation of RD(AK)-CFP/YFP (n=3). (* indicates a p-
value
<0.05, ** indicates a p-value <0.001, error bars represent the SEM).
[0021] FIG.
17 depicts images and graphs showing Tau-RD aggregates
transfer between cells and induce further aggregation. (A). HEK293 cells
transfected
with RD(AK)-YFP were co-cultured for 48 h with an equivalent number of cells
expressing RD(LM)-HA. Cells were fixed with 4% paraformaldehyde and
immunofluorescence/X-34 staining was performed. Multiple cells showed
colocalization
of RD(LM)-HA and RD(AK)-YFP within inclusions. These inclusions also stained
positive for X-34, indicating beta sheet structure (solid arrows). In
addition, some
RD(LM)-HA inclusions stained positive for X-34 but did not colocalize with
RD(AK)- YFP
inclusions (open arrow). (B). Two populations of cells, one expressing RD(AK)-
CFP/YFP, and the other expressing RD(LM)-HA, were co-cultured for 48 h. RD(PP)-
HA
or non-transfected cells, NT, were used as controls. FRET was increased by co-
culture
with RD(LM)-HA, but not with RD(PP)-HA, or mock-transfected cells (n=3). (C).
To test
for cell death induced by tau aggregates as a mechanism of tau release, HEK293
cells
were transfected for 48 h with RD-HA (PP, AK, or LM), or were mock-
transfected. Mock-
4

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
transfected cells were treated with varying concentrations of staurosporine
(1, 2, 4, 20
pM) for 30 minutes at 37 C to induce cell death. Cells were then exposed to 5
pg/ml of
propidium iodide and fluorescence was determined via plate reader. No evidence
for
cell death in the various transfected populations was observed. (** indicates
a p-value
<0.001, error bars represent the SEM).
[0022] FIG. 18 depicts images and graphs showing RD aggregates
propagate misfolding between cells. HEK293 cells were co-transfected with
various RD-
CFP and RD-HA constructs. 15 h later, these cells were co-cultured with cells
expressing RD(AK)-YFP or RD(PP)-YFP for 48 h (A) FRET microscopy was performed

to determine whether co-aggregation occurred via direct protein contact. CFP
signal
was measured before and after photobleaching of YFP. RD(LM)-CFP and RD(LM)-YFP

aggregates had a mean FRET efficiency of 14.2% 0.053 SD (n=11) indicative of

RD(LM)-CFP and RD(LM)-YFP in direct contact. The upper and lower panels depict
the
acceptor and donor channels, respectively, before (Pre) and after (Post)
photobleaching. A representative heat map of the calculated FRET efficiency is
shown
at top right. The histogram depicts the calculated FRET efficiency on a pixel-
by-pixel
basis. The FRET efficiency of Tau RD aggregate was -25% in this cell. Negative
values
are derived from unpaired CFP. (B) A FRET signal was observed when cells
expressing
RD(AK)-CFP/RD-HA were co-cultured with cells expressing RD(AK)-YFP. This
signal
increased when aggregation of RD(AK)-CFP was induced by co-expression of
aggregation-prone forms of tau, either AK, or LM mutants. No significant
signal was
noted when either RD-CFP or RD-YFP contained the PP mutation that blocks [3-
sheet
formation (n=3). (C) To test for amplification of misfolding, populations of
cells
expressing CFP alone or RD(LM)-CFP were preexposed for 48 h to cells
expressing
RD-HA with either PP, AK, or LM mutations to promote misfolding to varying
degrees.
These co-cultured populations were then split and co-cultured for 48 h with
cells
expressing RD(AK)-YFP to determine the degree of aggregation reported by cell-
cell
transfer and FRET. Prior exposure of RD(LM)-HA cells to the RD(AK)-CFP cell
population increased FRET signal by 2.6 fold vs. prior exposure to RD(PP)-HA .

Interposition of cells expressing pure CFP in the second population of cells
completely

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
blocked the effect of prior exposure to aggregation-prone RD-HA mutants (n=3).
(*
indicates a p-value <0.05, ** indicates a p-value <0.001, error bars represent
the SEM).
[0023] FIG. 19 depicts graphs and an immunoblot showing propagation
of
tau aggregates through the extracellular medium. (A) HEK293 cells transfected
with
RD(LM)-HA were co-cultured for 48 h with an equivalent number of RD(AK)-
CFP/YFP
cells prior to FRET analysis. Increasing the volume of cell culture medium
reduced the
efficiency of trans-cellular movement of aggregates. (B) Transfer of
conditioned medium
from cells expressing RD(LM)-HA to cells expressing RD(AK)-CFP/YFP was
sufficient
to induce aggregation by 60%. (C) HJ9.3 antibody added to the media reduced
FRET,
consistent with interference with propagation of aggregation. (D) Non-specific
IgG had
no effect on propagation. (E) HJ9.3 had no effect on intracellular aggregation
of
RD(AK)-CFP/YFP co-expressed within the same cell. (F) HJ9.3 blocked the effect
of
RD(LM)-HA to induce RD(AK)-YFP in co-cultured cells, as determined by
detergent
fractionation and western blot. (T. Total protein, S. Soluble protein and P.
Pellet
insoluble protein, (G) Quantitative analysis of three independent Western
blots revealed
a -60% decrease in the pellet fraction, relative to the total fraction, after
exposure to
HJ9.3. (H) Cells expressing RD(LM)-YFP and mCherry were co-cultured and
analyzed
by flow cytometry. HJ9.3 decreased the percentage of dual positive cells from
2.07% to
1.31 /o. Cells mixed just prior to cytometry were a background control (*
indicates a p-
value <0.05, ** indicates a p-value <0.001, error bars represent the SEM).
[0024] FIG. 20 depicts images of HEK293 cells transfected with
RD(AK) -
YFP (top panels) or mock-transfected (lower panels). HJ9.3 was added to the
culture
medium for the 48 h period. At the end of the experiment, the cells were
fixed,
permeabilized, and stained with an anti-mouse secondary antibody (labeled with
Alexa
546). Confocal microscopy was used to analyze the localization of HJ9.3/tau
complexes. The top panels show that many complexes are identified when RDA(K) -

YFP is expressed, but none in its absence (lower panels). Orthogonal analyses
(right
panel) demonstrate that most complexes are present at the cell surface,
although
occasional intracellular complexes were observed.
6

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0025] FIG. 21 depicts images and a graph showing Tau fibrils
mediate
cell-cell propagation. (A) Conditioned media was collected from transfected
cell
populations co-cultured for 0 h or 48 h with HJ9.3 or control IgG antibody
(1:1000),
followed by immunoprecipitation and Western blot. HJ9.3 specifically captured
tau RD
species from the cell media, while IgG did not. Higher-order aggregated
species were
present upon expression of RD(AK)- YFP or RD(LM)-YFP but not RD(PP)-YFP. (B)
Quantitative analyses of three independent Western blots showed a -10-fold
increase in
the tau after 48 h incubation. (C) Cells were exposed to HJ9.3 for various
times. (D)
Purified antibody/antigen complexes from media exposed for 48 h to HJ9.3 were
deposited on AFM chips for imaging. Obvious fibrillar species in the media of
cells
expressing RD(AK)-HA and RD(LM)-HA were observed, while RD(PP)-HA produced
only amorphous aggregates. Scale bar, lium.
[0026] FIG. 22 depicts a schematic and graphs showing HJ8.5 and
HJ9.4
activity against recombinant human tau. (A) depicts a schematic illustrating a
co-culture
of RD (LM)-CFP and RD(AK280)-YFP cells in presence and absence of different
monoclonal full length tau antibodies. (B) depicts a graph showing HJ8.5,
HJ9.3 and
HJ9.4 were able to block tau propagation. (C) depicts a graph showing HJ8.5,
HJ9.3
and HJ9.4 were able to detect RD-tau fibrils in an ELISA assay.
[0027] FIG. 23 depicts a schematic illustrating the experimental
plan for
(A) intracerebroventricular injection and (B) implantation of an osmotic pump
in the
lateral ventricle of each mouse. (C) shows an image verifying the placement of
the
cannula by cresyl violet staining.
[0028] FIG. 24 depicts images of the anti-tau antibodies after 6
weeks
infusion in P301S tg mice by (A) Coomassie blue staining and (B)
immunoblotting
against recombinant longest human tau isoform hTau40 using antibodies taken
from the
pump before and after 6 weeks infusion.
[0029] FIG. 25 depicts a graph showing lack of interference of
infused tau
antibodies in HJ8.7-BT2B ELISA for total tau. Indicated concentrations of
antibodies
were pre-incubated with recombinant human tau protein before applying to
ELISA.
7

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0030] FIG. 26 depicts images of coronal sections of piriform
cortex of
treated 9 month old P301S tg mice treated with vehicle/PBS (top panels) or
different
anti-tau monoclonal antibodies (HJ8.5, HJ9.3 as labeled in bottom panels).
Sections
were stained with biotinylated AT8 antibody, which recognizes an abnormally
phosphorylated form of tau.
[0031] FIG. 27 depicts graphs showing the percent of area covered
by
AT8 staining of neurofibrillary tangles in the (A) hippocampus CA2 and CA3,
(B)
amygdala, (C) piriform cortex, and (D) entorhinal cortex.
[0032] FIG. 28 graphs showing HJ9.3 antibody detection of tau
fibrils and
RD-tau monomer by ELISA. Different concentrations of RD-wt tau monomers and
fibrils
were coated on ELISA plate. HJ9.3 was used as the primary antibody. For the
detection
anti-mouse HRP linked antibody was used.
[0033] FIG. 29 depicts a schematic illustrating trans-cellular
propagation of
tau aggregation occurring via transfer of fibrils within the cell medium.
Protein aggregate
in a donor cell escapes the cell (A), enters a recipient cell (B), and
directly contacts
natively folded protein (C) to amplify the misfolded state (D). This cell-cell
movement is
mediated by fibrils that are released directly into the medium. These fibrils
can be
trapped within the extracellular space by an anti-tau antibody (HJ9.3) that
interferes with
cell-cell propagation (E).
[0034] FIG. 30. Characterization of anti-tau antibodies by surface
plasmon
resonance (SPR) and lmmunoblotting. The figure depicts SPR sensorgrams showing

the binding of each anti-tau antibody towards immobilized recombinant human
tau
(longest isoforms hTau40, 441 aa) and immobilized mouse tau (longest isoforms
mTau40, 432 aa). Each antibody was run with various concentrations (0.11,
0.23, 0.46,
0.90, 1.8, 3.7, 7.5 g/ml) and plots are shown in the corresponding color. (A)
SPR
sensorgrams of HJ9.3 antibody binding to immobilized human tau and immobilized

mouse tau (B). (C) SPR sensorgrams of HJ9.4 antibody binding to immobilized
human
tau and immobilized mouse tau (D). SPR sensorgrams of HJ8.5 antibody binding
to
immobilized (E) human and (F) mouse tau. (G) RAB soluble fractions of 3 month
old tau
knockout (KO), 3 month old wild-type (WT), 3 month old P3015 (3mo) and 9 month
old
8

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
P301S (9mo) mice were analyzed by immunoblot by using the indicated anti-tau
antibodies.
[0035] FIG. 31. SPR sensorgram of the interaction between anti-tau
antibodies towards immobilized human tau fibrils. SPR sensorgrams of HJ9.3
(A),
HJ9.4 (B) and HJ8.5 (C) anti tau antibodies run with various concentrations
towards
immobilized human tau fibrils.
[0036] FIG. 32. Characterization of anti-tau antibodies in
different assays.
lmmunostaining of brain sections from 3 month old tau knockout (KO), 3 month
old wild
type (WT), 3 month old P3015 (3mo), 12 month old P3015 (12mo) mice from the
region
of the piriform cortex and from the frontal cortex of Alzheimer's disease (AD)
tissue
were stained with biotinylated HJ8.5 antibody. Insert in 12 month old P301S
micrograph
shows cell body staining in addition to diffuse neuropil staining. Black arrow
indicates
the area magnified. Insert in human AD brain cortex micrograph shows the
staining of
neurofibrillary tangles (NFT) in higher magnification. Black arrow indicates
the area
magnified. Scale bar is 250 pm in panel with tau KO, same magnification
images. Scale
bar 50 pm in inserts of P301S 12mo and AD.
[0037] FIG. 33. Tau-antibodies block the uptake and seeding
activity of
P301S tau aggregates as detected by a FRET assay. H EK293 cells expressing
RD(AK280)-CFP/YFP were exposed to 2.5 pg of total protein of 1xTBS brain
lysates for
24 h. (A) Brain lysates collected from 12 mo old P3015 mice induced much
greater
seeding activity (n=5) as compared to lysates from knockout (KO) mice (n=7),
wild type
(WT) mice (n=6) or young 3-mo old P301S mice (n=2) (****p<0.0001). (B) HEK293
cells
were co-transfected with RD (AK280)-CFP and RD (AK280)-YFP. 18hrs later, pre-
incubated P3015 brain lysates with or without incubation of anti-tau
antibodies (HJ8.5,
HJ9.3 and HJ9.4) or control antibody (HJ3.4, anti Ar3 antibody) were added to
cells. All
the tau antibodies incubated with P3015 brain lysates significantly blocked
seeding
activity. Statistical significance was determined by one-way ANOVA followed by

Dunnett's post hoc test for multiple comparisons by using GraphPad Prism 5.0
software
(***p>0.001). (C) Titration of these antibodies with various concentrations
(0.125 pg/ml,
0.25 pg/ml, 0.5 pg/ml, 1 pg/ml and 2 pg/ml) was performed with a fixed amount
of
9

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
P301S brain lysates. 24 hrs later, FRET analysis was performed. Out of all tau-

antibodies we used, HJ8.5 was the most potent in blocking the uptake and
seeding
activity of P3015 brain lysates. Statistical significance was determined by
two-way
ANOVA followed by Bonferroni post hoc test for multiple comparisons. (** p <
0.0001, *
p < 0.01, Values represent mean SEM).
[0038] FIG. 34. No detected cellular uptake of tau antibodies bound
to
P3015 Tau aggregates. P3015 brain lysates were added to HEK293 cells for 3hrs.
For
detection of tau, all 3 different anti-tau or control (HJ3.4, Ar3 antibody)
antibodies were
used followed by Alexa-fluor546 anti-mouse IgG staining. In addition, P301S
brain
lysates were pre-incubated with and without 3 different anti-tau antibodies
and HJ3.4
antibody, then added to HEK293 cells, fixed and permeabilized. Alexa-fluor546
anti-
mouse IgG were used to identify the internalized antibodies. 4',6'-diamidino-2-

phenylindole (DAPI; shown in blue) was used for nuclear stain.
[0039] FIG. 35. Experimental outline of ICV infusion of antibodies
and
efficacy of antibody by different treatment method. (A) Experimental plan for
infusion of
antibodies or vehicle (PBS) by intracerebroventricular injection into the left
lateral
ventricle of the brain. (B) Representative cresyl violet staining of the
coronally sectioned
brain region to verify the surgically implanted probe placement into the left
lateral
ventricle. In this study, we included the mice which had correct probe
placements into
the left lateral ventricle.
[0040] FIG. 36. Anti-tau antibodies strongly decreased AT8 staining
in
P301S mouse brain. Representative coronal sections of PBS (A), HJ3.4 antibody
(B),
HJ8.5 antibody (C), HJ9.3 antibody (D) and HJ9.4 antibody (E) treated 9 month
old
P301S mice stained with biotinylated AT8 antibody in regions including the
piriform
cortex and amygdala. Scale bar is 250 m. Inserts in A to E show the higher
magnification of biotinylated AT8 antibody staining of phosphorylated tau,
scale bar is
50 m.
[0041] FIG. 37. Certain anti-tau antibodies strongly decrease AT8
staining
in P301S mouse brain. Percent of the area covered by biotinylated AT8 staining
of
abnormally phosphorylated tau in piriform cortex (A), entorhinal cortex (B),
amygdala

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
(C) and hippocampus CA1 region (D) in mice treated with the anti-tau
antibodies HJ8.5
(N=13), HJ9.3 (N=15), HJ9.4 (N=13), the anti-An antibody, HJ3.4 (N=8), or PBS
(N=16)
in 9 month old P3015 mice. There was reduced AT8 staining in several different
brain
regions in the anti-tau antibody treated mice compared to PBS or HJ3.4
antibody
treated mice. HJ8.5 had the largest effects. ** p < 0.01, * p < 0.05, values
represent
mean SEM.
[0042] FIG. 38. Quantification of biotinylated AT8 antibody
staining in male
and female P3015 mice. Percent of area covered by biotinylated AT8 staining of

abnormally phosphorylated tau in male (A) and female P3015 mice (B) in
piriform
cortex (A and E), entorhinal cortex (B and F), amygdala (C and G) and
hippocampal
CA1 regions (D and H) in anti-tau antibody (HJ8.5, HJ9.3 and HJ9.4), control
antibody
(HJ3.4) plus PBS treated mice.
[0043] FIG. 39. Some anti-tau antibodies strongly decrease ThioS
staining
of neurofibrillary tangles in P301S mouse brain. (A) Representative images of
ThioS
staining of neurofibrillary tangles in the piriform cortex of 9 month old
P301S mice
treated for 3 months with PBS, HJ3.4, HJ8.5, HJ9.3 and HJ9.4 antibodies. ThioS

staining of neurofibrillary tangles was reduced in HJ8.5antibody treated mice
compared
to the PBS or HJ3.4 antibody treated mice. Scale bar represents 100 m. (B)
Semi
quantitative assessment of ThioS staining by scoring from 1 (no staining) to 5

(maximum staining) in all anti-tau antibody and control treated mice. HJ8.5
antibody
treated mice had significantly less ThioS staining compared to PBS or HJ3.4
antibody
treated mice. *p<0.05, **p<0.01.
[0044] FIG. 40. Correlations between phospho-tau staining, and
activated
microglial staining. (A) Biotinylated AT8 staining of phospho-tau in HJ8.5
(N=6), HJ9.3
(N=6) and PBS treated 9 month old P301S mice (N=6 per each group) showed
strong
correlation with PHF1 staining, another phospho-tau antibody. (B) Strong
correlation
was observed between CD68 staining of activated microglia and biotinylated AT8

staining of phospho-tau in all groups (N=6 per each group) (C) lmmunoblotting
of
representative 70% FA fraction samples (N=4) were analyzed with polyclonal
mouse
anti-tau antibodies (Abcam).
11

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0045] FIG. 41. CD68 staining of activated microglia. Mice were
assessed
for microglial activation in P301S mice. Representative images of CD68
staining of
activated microglia in the piriform cortex of 9 month old P301S mice treated
with PBS
(A), HJ3.4 antibody (B), HJ8.5 antibody (C), HJ9.3 antibody (D) and HJ9.4
antibody (E).
[0046] FIG. 42. Insoluble tau levels are reduced by antibodies
HJ8.5 and
HJ9.3 in P3015 mice. The cortex of all the treated mice [PBS (N=16), HJ3.4
antibody
(N=8) HJ8.5 (N=13), HJ9.3 (N=15), HJ9.4 (N=13)] were sequentially extracted by
RAB
(A), RI PA (B) and 70% FA (C) and their tau levels were quantified by ELISA.
There
were no statistical differences in soluble tau levels in RAB and RI PA
fractions between
the groups. However, there was a significant decrease of insoluble tau levels
in 70% FA
fractions in the HJ8.5 and HJ9.3 anti-tau antibodies treated mice compared to
the PBS
or HJ3.4 antibody treated groups. Insoluble tau levels in the HJ9.4 antibody
treated
mice were not different from the control groups (**p<0.01). Levels of human
tau (D),
mouse tau (E) and phospho tau at Ser202 and Thr205 (F) levels were assessed in
70%
FA fractions by specific anti-human, anti-mouse, or anti-phospho tau
antibodies by
ELISA (n=6 mice per treatment group). There was a decrease in human tau levels
in all
groups of anti-tau antibody treated mice and no change in mouse tau levels. In
70% FA
fractions, we also found that phospho tau at Ser202 and Thr205 as detected by
AT8
reactivity was reduced in anti-tau antibody treated mice compared to controls,
similar to
total human tau.
[0047] FIG. 43. Anti-tau antibody treated P3015 mice have decreased
tau
seeding activity in cortical extracts as detected by FRET assay. (A) Tau
seeding activity
was measured with RAB soluble fractions of all PBS (N=16), HJ3.4 (N=8), HJ8.5
(N=13), HJ9.3 (N=15), and HJ9.4 (N=13) treated mice on HEK293 cells by FRET
assay.
HEK293 cells were co-transfected with RD (AK280)-CFP and RD (AK280)-YFP. 18
hrs
later, RAB soluble fractions were added to cells. Seeding activity was
significantly
reduced in HJ8.5, and HJ9.3 antibody treated mice compared to the PBS or HJ3.4

antibody treated mice. RAB soluble fractions from HJ9.4 antibody treated mice
did not
have decreased seeding activity compared to the PBS or HJ3.4 antibody RAB
soluble
fractions (***p<0.001, Values represent mean SEM). (B) RAB soluble fractions
were
12

CA 02877397 2014-12-18
WO 2014/008404
PCT/US2013/049333
immunoprecipitated from tau knockout, PBS, or anti-tau antibody treated mice.
Elution
of any seeding activity from the antibody/bead complexes was measured by FRET
assay. There was significantly less seeding activity observed in HJ8.5 and
HJ9.3
antibody treated mice versus PBS-treated mice (****p<0.0001, values represent
mean
SEM). (C) 70% FA fractions of 9 month old P3015 brain cortex region of all
treated
groups analyzed by ELISA showed a strong correlation with FRET analysis
performed
with the RAB soluble fractions. (D) Comparison between tau levels (X-axis) and
seeding
activity (Y-axis) present in RAB soluble fractions of 9 month old P3015 brain
cortex of
all treated mice assessed. There was no significant correlation between these
2
measures. (E) Tau species in the RAB soluble fractions of 3 month old knockout
(KO), 3
month old wild type (WT), 3 month old P3015, and 9 month old PBS-treated P3015

mice were separated on SDD-AGE, followed by western blotting. Polyclonal mouse
anti-
tau antibody was used for detecting tau species. High molecular weight tau
species
present in the RAB soluble fraction in both 3 month old P3015 mice and larger
amounts
present in 9 month old P301S mice.
[0048] FIG.
44. Groups did not differ significantly in terms of locomotor
activity, sensorimotor function or on the auditory cue component of the
conditioned fear
test. The results of rmANOVAs failed to reveal significant main or interaction
effects
involving Treatment for total ambulations in the holeboard test (A), for the
ledge test (B)
or any other of the sensorimotor measures (not shown), or on the accelerating
rotarod
(C). Data from the altered context baseline on day 3 of conditioned fear
testing yielded a
significant effect of Treatment (*p=0.027) and subsequent comparisons showed
that a
large portion of this effect was due to significant differences between the
HJ9.4 mice
and the PBS+HJ3.4 control group (p=0.0007). (D). However, no significant main
or
interaction effects of Treatment were found following an rmANOVA on the
auditory cue
data (min 3-10) suggesting that the freezing levels were not significantly
different among
the groups during this time (E). To assess whether activity levels may have
had an
effect on freezing during the contextual fear test on day 2, we computed
Pearson's
correlation coefficient (r) between total ambulations measured during the
holeboard test
13

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
versus % time spent freezing during the contextual fear test and found that
they were
not significantly correlated (p=0.39) (F).
[0049] FIG. 45. Contextual fear conditioning deficits in P301S tau
transgenic mice are rescued by HJ8.5 and HJ9.4 antibody treatments. (A) On day
1 of
conditioned fear testing, no differences were observed among groups in
freezing levels
during either the 2-min baseline condition or the tone/shock (T/S) training as
indicated
by the lack of a significant main or interaction effects involving Treatment
following
rmANOVAs on these data. (B) In contrast, a significant effect of Treatment
(*p=0.019)
and a significant Treatment by Minutes interaction (**p=0.0001) were observed
following
an rmANOVA on freezing levels during the contextual fear testing on day 2.
Only the
HJ9.4 group showed significant habituation from minute 1 versus minute 8,
(#p=0.002).
(C) Subsequent planned comparisons showed that freezing in the HJ8.5 and HJ9.4
tau
antibody groups was significantly increased relative to the PBS+HJ3.4 control
group
when averaged across the 8-min session (**p=0.006 and *p=0.022, respectively).

However, further analyses of the data showed that the largest differences
between the
HJ9.4 group and the PBS+HJ3.4 controls occurred during minute 2 (tp=0.004),
while
the largest differences between the HJ8.5 treated mice and the control group
were
found during minutes 4-7 (ttp<0.004) as depicted in "B".
[0050] FIG. 46 depicts a graph showing a sandwich Tau ELISA assay
can
be used to discriminate between plasma samples that are positive for seeding
activity
and plasma samples that are negative for seeding activity. Seeding activity
was
determined as described in Kfoury et al 2012 J Biol Chem 287(23). Amount of
tau
aggregate is reported as relative fold-change induction over signal from
plasma
collected from healthy young humans (i.e. background signal of the assay).
[0051] FIG. 47 depicts graphs showing the effect of anti-tau
antibodies of
the invention on a tau cellular propagation assay. In each graph, the first
bar represents
medium without added antibody, representing baseline efficiency of
propagation. (A)
HJ8.1 and HJ8.2; (B) HJ8.3 and HJ8.4; (C) HJ8.5 and HJ8.7; (D) HJ8.8 and
HJ9.1; (E)
HJ9.2 and HJ9.3; (F) HJ9.4 and HJ9.5.
14

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0052] FIG. 48 depicts a graph showing the effect on tau
propagation of
individual anti-tau antibodies or equimolar mixtures of anti-tau antibodies in
a cell-based
assay.
[0053] FIG. 49 depicts in (A) a graph showing HJ9.3 antibody has no
effect on intracellular tau aggregation when RD(AK)-CFP/YFP are co-expressed
within
the same cell, and in (B) a graph showing that nonspecific IgG has no effect
on trans-
cellular propagation of tau aggregation.
[0054] FIG. 50 depicts a graph showing HJ9.3 inhibits tau aggregate
uptake, as measured by flow cytometry. Cells were exposed to recombinant RD
fibrils
that were chemically labeled with a fluorescent dye. After trypsinization and
dispersion,
the cells were counted using a flow cytometer. HJ9.3 dose-dependently reduces
the
number of fluorescently labeled cells, indicating inhibition of aggregate
uptake.
DETAILED DESCRIPTION
[0055] The common minimal connection between Alzheimer's Disease
and all the tauopathies is the aggregation state of tau. Under all these
diseased
conditions, monomeric tau is known to be converted into polymeric ordered
fibrils.
Neurofibrillary tangles (NFTs), which are comprised of fibrillar tau
aggregates, are a
neuropathological hallmark of tauopathies. Applicants have discovered that
spreading of
tau pathology in the brain may be caused by a form of tau aggregate released
from a
"donor" cell entering a second "recipient" cell, and inducing further
misfolding and
aggregation of tau in the recipient cell via direct protein-protein contact.
The specific
form of tau aggregate which facilitates this cell-to-cell spread of tau
aggregates is
referred to as "tau seeds" and the activity may be referred to herein as
"seeding
activity", since this form of tau aggregate seeds or nucleates tau aggregation
in the cell
it enters (i.e. the "recipient cell").
[0056] Tau can exist in both a monomeric form and in different
aggregated
forms. As used herein, the term "tau aggregate" refers to a molecular complex
that
comprises two or more tau monomers. Without wishing to be bound by theory, a
tau
aggregate may comprise a nearly unlimited number of monomers bound together.
For

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
example, a tau aggregate may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10 or more tau
monomers.
Alternatively, a tau aggregate may comprise 20, 30, 40, 50, 60, 70, 80, 90,
100 or more
tau monomers. A tau aggregate may also comprise 500, 1000, 1500, 2000, 2500,
3000,
3500, 4000, 4500, 5000 or more tau monomers. The terms "fibrillar tau
aggregate" and
"tau fibril" refer to forms of tau aggregates, and these terms are used
interchangeably
herein. A fibrillar tau aggregate is a polymeric, ordered fiber comprising
tau. Tau fibrils
are generally not soluble, but shorter assemblies, or oligomers, can be
soluble. Tau
aggregate also refers to soluble tau oligomers and protofibrils, which may act
as
intermediates during tau aggregation. Also included in the definition of tau
aggregate is
the term "tau seed", which refers to a tau aggregate that is capable of
nucleating or
"seeding" intracellular tau aggregation when internalized by a cell, or when
exposed to
monomeric tau in vitro. Tau seeding activity may be assessed in a cellular tau

aggregation assay as described herein.
[0057] In addition, applicants have discovered antibodies that
specifically
bind to tau and methods of use thereof. In an aspect, the present invention
provides
antibodies that specifically bind tau. In another aspect, the present
invention provides
means for effectively slowing and/or reducing cell-to-cell propagation of tau
aggregation.
Antibodies of the invention may slow and/or reduce the propagation of tau
aggregation
by promoting the disaggregation of protein fibrils, blockading the conversion
of
monomeric tau into aggregated tau in the cell, promoting intracellular
degradation of tau
aggregates, preventing entry of the tau aggregates into neighboring cells, or
a
combination thereof. In another aspect, the present invention provides means
to detect
tau aggregate in a sample of biological fluid obtained from a subject. In
another aspect,
the present invention provides means to measure the amount of tau aggregate in
a
sample of biological fluid obtained from a subject. In another aspect, the
present
invention provides means to classify a subject based on the amount of tau
aggregate
measured in a sample of biological fluid obtained from a subject. Classifying
a subject
based on the amount of tau aggregate measured in a sample of biological fluid
obtained
from the subject may be used to identify subjects that will develop a symptom
and/or
disease associated with tau aggregation in the subject's lifetime.
16

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0058] The present invention encompasses the discovery that anti-
tau
antibodies may slow the propagation of fibrillar tau aggregates by binding
extracellular
tau released from cells, thereby preventing entry of the tau aggregates into
neighboring
cells and slowing spread of tau aggregation. In an aspect, the present
invention
provides means for preventing entry of a tau aggregate into a cell. In another
aspect,
the present invention provides means for reducing intracellular tau
aggregation. In
another aspect, the present invention provides means for decreasing tau
seeding
activity. Antibodies of the invention useful in preventing entry of the tau
aggregates into
neighboring cells include those which bind an epitope within tau.
I. Antibodies that bind to tau
[0059] In humans, there are six isoforms of tau that are generated
by
alternative splicing of exons 2, 3, and 10. The isoforms ranging from 352 to
441 amino
acids. Exons 2 and 3 encode 29-amino acid inserts each in the N-terminus
(called N,
and hence, tau isoforms may be 2N (both inserts), 1N (exon 2 only), or ON
(neither). All
tau isoforms have three repeats of the microtubule binding domain. Inclusion
of exon 10
at the C-terminus leads to inclusion of a fourth microtubule binding domain
encoded by
exon 10. Hence, tau isoforms may be comprised of four repeats of the
microtubule
binding domain (exon 10 included) or three repeats of the microtubule binding
domain
(exon 10 excluded). Anti-tau antibodies of the invention may include
antibodies that bind
any of the isoforms of tau. In an exemplary embodiment, anti-tau antibodies of
the
invention may include antibodies that bind to an isoform of tau that comprises
exon 10.
[0060] As noted above, tau can be found in soluble and insoluble
compartments, in monomeric and aggregated forms, in ordered or disordered
structures, intracellularly and extracellularly, and may be complexed with
other proteins
or molecules. Anti-tau antibodies of the invention may include antibodies that
bind to
one or more forms of tau as described. In some embodiments, an anti-tau
antibody
binds a tau monomer. In other embodiments, an anti-tau antibody binds a tau
aggregate. In still other embodiments, an anti-tau antibody binds a tau
fibril. In different
embodiments, an anti-tau antibody binds a tau monomer and a tau aggregate. In
17

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
alternative embodiments, an anti-tau antibody binds to a tau aggregate and a
tau fibril.
In different embodiments, an anti-tau antibody binds to a tau fibril and a tau
monomer.
[0061] Anti-tau antibodies useful herein also include all
antibodies that
specifically bind tau aggregates present in a biological sample. Anti-tau
antibodies
useful herein also include all antibodies that reduce cell-to-cell propagation
of tau
aggregation. In other words, useful antibodies slow and/or decrease the amount
of tau
that enters recipient cells, compared to the amount that would enter a
recipient cell in
the absence of an antibody of the invention. Hence, useful antibodies decrease
the
amount of tau aggregation that occurs in the recipient cells.
[0062] In an aspect, antibodies useful herein include those
antibodies
which have been isolated, characterized, purified, are functional and have
been
recovered (obtained) for use in a functional therapeutic composition which is
administered to a living subject. In another aspect, antibodies useful herein
include
those antibodies which have been isolated, characterized, purified, are
functional and
have been recovered (obtained) for use in an assay to detect tau aggregates in
a
biological sample obtained from a living subject and predict the development
of
symptoms associated with tau aggregation over the lifetime of the subject. In
another
aspect, antibodies useful herein include those antibodies which have been
isolated,
characterized, purified, are functional and have been recovered (obtained) or
for use in
an assay to detect tau aggregates in a biological sample obtained from a
living subject
and classify the subject as having an increased risk of developing symptoms
associated
with tau aggregation over the subject's lifetime. In another aspect,
antibodies useful
herein include those antibodies which have been isolated, characterized,
purified, are
functional and have been recovered (obtained) for use and are listed in Table
A, as well
as variants thereof (e.g. humanized forms, chimeric forms, and immunological
fragments).
18

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Table A. Antibodies of the invention
Antibody Name Tau epitope
HJ8.1.1 DRKDQGGYTMHQD (SEQ ID NO: 1)
HJ8.1.2 TDHGAE (SEQ ID NO: 10)
HJ8.2 PRHLSNV (SEQ ID NO: 3)
HJ8.3 PRHLSNV (SEQ ID NO: 3)
HJ8.4 KTDHGA (SEQ ID NO: 11)
HJ8.5 DRKDQGGYTMHQD (SEQ ID NO: 1)
HJ8.7 AAGHV (SEQ ID NO: 5)
HJ8.8 EPRQ (SEQ ID NO: 4)
HJ9.1 TDHGAEIVYKSPVVSG (SEQ ID NO: 6)
HJ9.2 EFEVMED (SEQ ID NO: 7)
HJ9.3 GGKVQIINKK (SEQ ID NO: 8)
HJ9.4 EFEVMED (SEQ ID NO: 7)
HJ9.5 EFEVMED (SEQ ID NO: 7)
[0063] The term "antibody" includes the term "monoclonal antibody".
"Monoclonal antibody" refers to an antibody that is derived from a single copy
or clone,
including e.g., any eukaryotic, prokaryotic, or phage clone. "Monoclonal
antibody" is not
limited to antibodies produced through hybridoma technology. Monoclonal
antibodies
can be produced using e.g., hybridoma techniques well known in the art, as
well as
recombinant technologies, phage display technologies, synthetic technologies
or
combinations of such technologies and other technologies readily known in the
art.
Furthermore, the monoclonal antibody may be labeled with a detectable label,
immobilized on a solid phase and/or conjugated with a heterologous compound
(e.g., an
enzyme or toxin) according to methods known in the art.
[0064] Further by "antibody" is meant a functional monoclonal
antibody, or
an immunologically effective fragment thereof; such as an Fab, Fab', or
F(ab')2
fragment thereof. In some contexts herein, fragments will be mentioned
specifically for
emphasis; nevertheless, it will be understood that regardless of whether
fragments are
19

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
specified, the term "antibody" includes such fragments as well as single-chain
forms.
As long as the protein retains the ability specifically to bind its intended
target, it is
included within the term "antibody." Also included within the definition
"antibody" for
example are single chain forms, generally designated Fv regions, of antibodies
with this
specificity. Preferably, but not necessarily, the antibodies useful in the
discovery are
produced recombinantly, as manipulation of the typically murine or other non-
human
antibodies with the appropriate specificity is required in order to convert
them to
humanized form. Antibodies may or may not be glycosylated. Antibodies are
properly
cross-linked via disulfide bonds, as is known.
[0065] The basic antibody unit of an antibody useful herein
comprises a
tetramer. Each tetramer is composed of two identical pairs of polypeptide
chains, each
pair having one "light' (about 25 kDa) and one "heavy" chain (about 50-70
kDa). The
amino-terminal portion of each chain includes a variable region of about 100
to 110 or
more amino acids primarily responsible for antigen recognition. The carboxy-
terminal
portion of each chain defines a constant region primarily responsible for
effector
function.
[0066] Anti-tau antibodies useful herein include those which are
isolated,
characterized, purified, function and have been recovered (obtained) from a
process for
their preparation and thus available for use herein in a useful form in a
therapeutically,
medicinally, or diagnostically sufficient amount.
[0067] Light chains are classified as gamma, mu, alpha, and lambda.
Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define
the
antibody's isotype as lg0, IgM, IgA, IgD and IgE, respectively. Within light
and heavy
chains, the variable and constant regions are joined by a "J" region of about
12 or more
amino acids, with the heavy chain also including a "D" region of about 10 more
amino
acids.
[0068] The variable regions of each light/heavy chain pair form the
antibody binding site. Thus, an intact antibody has two binding sites. The
chains exhibit
the same general structure of relatively conserved framework regions (FR)
joined by
three hypervariable regions, also called complementarily determining regions

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
(hereinafter referred to as "CDRs.") The CDRs from the two chains are aligned
by the
framework regions, enabling binding to a specific epitope. From N-terminal to
C-
terminal, both light and heavy chains comprise the domains FR1, CDR1, FR2,
CDR2,
FR3, CDR3 and FR4 respectively. The assignment of amino acids to each domain
is in
accordance with known conventions (See, Kabat "Sequences of Proteins of
Immunological Interest" National Institutes of Health, Bethesda, Md., 1987 and
1991;
Chothia, et al, J. Mol. Bio. (1987) 196:901-917; Chothia, et al., Nature
(1989) 342:878-
883).
[0069] In an aspect, monoclonal anti-tau antibodies are generated
with
appropriate specificity by standard techniques of immunization of mammals,
forming
hybridomas from the antibody-producing cells of said mammals or otherwise
immortalizing them, and culturing the hybridomas or immortalized cells to
assess them
for the appropriate specificity. In the present case, such antibodies could be
generated
by immunizing a human, rabbit, rat or mouse, for example, with a peptide
representing
an epitope encompassing a region of the tau protein coding sequence or an
appropriate
subregion thereof. Materials for recombinant manipulation can be obtained by
retrieving
the nucleotide sequences encoding the desired antibody from the hybridoma or
other
cell that produces it. These nucleotide sequences can then be manipulated and
isolated, characterized, purified and, recovered to provide them in humanized
form, for
use herein if desired.
[0070] As used herein "humanized antibody" includes an anti-tau
antibody
that is composed partially or fully of amino acid sequences derived from a
human
antibody germline by altering the sequence of an antibody having non-human
complementarity determining regions ("CDR"). The simplest such alteration may
consist
simply of substituting the constant region of a human antibody for the murine
constant
region, thus resulting in a human/murine chimera which may have sufficiently
low
immunogenicity to be acceptable for pharmaceutical use. Preferably, however,
the
variable region of the antibody and even the CDR is also humanized by
techniques that
are by now well known in the art. The framework regions of the variable
regions are
substituted by the corresponding human framework regions leaving the non-human
21

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
CDR substantially intact, or even replacing the CDR with sequences derived
from a
human genome. CDRs may also be randomly mutated such that binding activity and

affinity for tau is maintained or enhanced in the context of fully human
germline
framework regions or framework regions that are substantially human.
Substantially
human frameworks have at least 90%, 95%, or 99% sequence identity with a known

human framework sequence. Fully useful human antibodies may also be produced
in
genetically modified mice whose immune systems have been altered to correspond
to
human immune systems. As mentioned above, it is sufficient for use in the
methods of
this discovery, to employ an immunologically specific fragment of the
antibody, including
fragments representing single chain forms.
[0071] Further, as used herein the term "humanized antibody" refers
to an
anti-tau antibody comprising a human framework, at least one CDR from a
nonhuman
antibody, and in which any constant region present is substantially identical
to a human
immunoglobulin constant region, i.e., at least about 85-90%, preferably at
least 95%
identical. Hence, all parts of a humanized antibody, except possibly the CDRs,
are
substantially identical to corresponding pairs of one or more native human
immunoglobulin sequences.
[0072] If desired, the design of humanized immunoglobulins may be
carried out as follows. When an amino acid falls under the following category,
the
framework amino acid of a human immunoglobulin to be used (acceptor
immunoglobulin) is replaced by a framework amino acid from a CDR-providing
nonhuman immunoglobulin (donor immunoglobulin): (a) the amino acid in the
human
framework region of the acceptor immunoglobulin is unusual for human
immunoglobulin
at that position, whereas the corresponding amino acid in the donor
immunoglobulin is
typical for human immunoglobulin at that position; (b) the position of the
amino acid is
immediately adjacent to one of the CDRs; or (c) any side chain atom of a
framework
amino acid is within about 5-6 angstroms (center-to-center) of any atom of a
CDR
amino acid in a three dimensional immunoglobulin model (Queen, et al., op.
cit., and
Co, ct al, Proc. Natl. Acad. Sci. USA (1991) 88:2869). When each of the amino
acids in
the human framework region of the acceptor immunoglobulin and a corresponding
22

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
amino acid in the donor immunoglobulin is unusual for human immunoglobulin at
that
position, such an amino acid is replaced by an amino acid typical for human
immunoglobulin at that position.
[0073] In all instances, an antibody of the invention specifically
binds tau.
In exemplary embodiments, an antibody of the invention specifically binds
human tau.
The phrase "specifically binds" herein means antibodies bind to the protein
with an
affinity constant or Affinity of interaction (KD) in the range of 0.1 pM to 10
nM, with a
preferred range being 0.1 pM to 1 nM. The sequence of tau from a variety of
species is
known in the art, and methods of determining whether an antibody binds to tau
are
known in the art. For instance, see the Examples.
[0074] The antibodies of the present invention may also be used as
fusion
proteins known as single chain variable fragments (scFv). These scFvs are
comprised
of the heavy and light chain variable regions connected by a linker. In most
instances,
but not all, the linker may be a peptide. A linker peptide is preferably from
about 10 to
25 amino acids in length. Preferably, a linker peptide is rich in glycine, as
well as serine
or theronine. ScFvs can be used to facilitate phage display or can be used for
flow
cytometry, immunohistochemistry, or as targeting domains. Methods of making
and
using scFvs are known in the art.
[0075] In a preferred embodiment, the scFvs of the present
invention are
conjugated to a human constant domain. In some embodiments, the heavy constant

domain is derived from an IgG domain, such as IgG1, IgG2, IgG3, or IgG4. In
other
embodiments, the heavy chain constant domain may be derived from IgA, IgM, or
IgE.
[0076] An isolated antibody of the present invention that binds to
tau
preferably recognizes one of several epitopes. In one embodiment, the isolated

antibody of the present invention that binds to tau recognizes an epitope
listed in Table
A. In another embodiment, the isolated antibody of the present invention that
binds to
tau recognizes an epitope within the amino acid sequences of SEQ ID NO: 1
(DRKDQGGYTMHQD). Preferably, the isolated antibody recognizes an epitope
within
at least three contiguous amino acids of SEQ ID NO: 1, including within at
least 6
contiguous amino acids of SEQ ID NO: 1, within at least 7 contiguous amino
acids of
23

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
SEQ ID NO: 1, within at least 8 contiguous amino acids of SEQ ID NO: 1, within
at least
9 contiguous amino acids of SEQ ID NO: 1, within at least 10 contiguous amino
acids of
SEQ ID NO: 1, within at least 11 contiguous amino acids of SEQ ID NO: 1,
within at
least 12 contiguous amino acids of SEQ ID NO: 1, and within at least 13
contiguous
amino acids of SEQ ID NO: 1. In an exemplary embodiment, an isolated antibody
of the
present invention that recognizes an epitope within SEQ ID NO: 1 is the
antibody HJ8.5.
In another exemplary embodiment, an isolated antibody of the present invention
that
recognizes an epitope within SEQ ID NO: 1 is the antibody HJ8.1.1.
[0077] In another embodiment, the isolated antibody of the present
invention that binds to tau recognizes an epitope within the amino acid
sequence of
SEQ ID NO: 2 (KTDHGAE). Preferably, the isolated antibody recognizes an
epitope
within at least three contiguous amino acids of SEQ ID NO: 2, including within
at least 4
contiguous amino acids of SEQ ID NO: 2 within at least 5 contiguous amino
acids of
SEQ ID NO: 2 within at least 6 contiguous amino acids of SEQ ID NO: 2, and
within at
least 7 contiguous amino acids of SEQ ID NO: 2. In an exemplary embodiment, an

isolated antibody of the present invention that recognizes an epitope within
SEQ ID NO:
2 is the antibody HJ8.1.2. In another exemplary embodiment, an isolated
antibody of the
present invention that recognizes an epitope within SEQ ID NO: 2 is the
antibody HJ8.4.
[0078] In another embodiment, the isolated antibody of the present
invention that binds to tau recognizes an epitope within the amino acid
sequence of
SEQ ID NO: 3 (PRHLSNV). Preferably, the isolated antibody recognizes an
epitope
within at least three contiguous amino acids of SEQ ID NO: 3, including within
at least 4
contiguous amino acids of SEQ ID NO: 3, within at least 5 contiguous amino
acids of
SEQ ID NO: 3, within at least 6 contiguous amino acids of SEQ ID NO: 3, and
within at
least 7 contiguous amino acids of SEQ ID NO: 3. In an exemplary embodiment, an

isolated antibody of the present invention that recognizes an epitope within
SEQ ID NO:
3 is the antibody HJ8.2. In another exemplary embodiment, an isolated antibody
of the
present invention that recognizes an epitope within SEQ ID NO: 3 is the
antibody HJ8.3.
[0079] In still another embodiment, the isolated antibody of the
present
invention that binds to tau recognizes an epitope within the amino acid
sequences of
24

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
SEQ ID NO: 4 (EPRQ). Preferably, the isolated antibody recognizes an epitope
within at
least three contiguous amino acids of SEQ ID NO: 4, including within at least
4
contiguous amino acids of SEQ ID NO: 4. In an exemplary embodiment, an
isolated
antibody of the present invention that recognizes an epitope within SEQ ID NO:
4 is the
antibody HJ8.8.
[0080] In yet a further embodiment, the isolated antibody of the
present
invention that binds to tau recognizes an epitope within the amino acid
sequence of
SEQ ID NO: 5 (AAGHV). Preferably, the isolated antibody recognizes an epitope
within
at least three contiguous amino acids of SEQ ID NO: 5, including within at
least 4
contiguous amino acids of SEQ ID NO: 5, and within at least 5 contiguous amino
acids
of SEQ ID NO: 5. In an exemplary embodiment, an isolated antibody of the
present
invention that recognizes an epitope within SEQ ID NO: 5 is the antibody
HJ8.7.
[0081] In an additional embodiment, the isolated antibody of the
present
invention that binds to tau recognizes an epitope within the amino acid
sequence of
SEQ ID NO: 6 (TDHGAEIVYKSPVVSG). Preferably, the isolated antibody recognizes
an epitope within at least five contiguous amino acids of SEQ ID NO: 6,
including within
at least 6 contiguous amino acids of SEQ ID NO: 6, within at least 7
contiguous amino
acids of SEQ ID NO: 6, within at least 8 contiguous amino acids of SEQ ID NO:
6, within
at least 9 contiguous amino acids of SEQ ID NO: 5, within at least 9
contiguous amino
acids of SEQ ID NO: 6, within at least 10 contiguous amino acids of SEQ ID NO:
6,
within at least 11 contiguous amino acids of SEQ ID NO: 6, within at least 12
contiguous
amino acids of SEQ ID NO: 6, within at least 13 contiguous amino acids of SEQ
ID NO:
6, within at least 14 contiguous amino acids of SEQ ID NO: 6, within at least
15
contiguous amino acids of SEQ ID NO: 6, and within at least 16 contiguous
amino acids
of SEQ ID NO: 6. In an exemplary embodiment, an isolated antibody of the
present
invention that recognizes an epitope within SEQ ID NO: 6 is the antibody
HJ9.1.
[0082] In another embodiment, the isolated antibody of the present
invention that binds to tau recognizes an epitope within the amino acid
sequence of
SEQ ID NO: 7 (EFEVMED). Preferably, the isolated antibody recognizes an
epitope
within at least three contiguous amino acids of SEQ ID NO: 7, including within
at least 4

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
contiguous amino acids of SEQ ID NO: 6, within at least 5 contiguous amino
acids of
SEQ ID NO: 7, within at least 6 contiguous amino acids of SEQ ID NO: 7, and
within at
least 7 contiguous amino acids of SEQ ID NO: 7. In an exemplary embodiment, an

isolated antibody of the present invention that recognizes an epitope within
SEQ ID NO:
7 is the antibody HJ9.2. In an exemplary embodiment, an isolated antibody of
the
present invention that recognizes an epitope within SEQ ID NO: 7 is the
antibody HJ9.4.
In an exemplary embodiment, an isolated antibody of the present invention that

recognizes an epitope within SEQ ID NO: 7 is the antibody HJ9.5.
[0083] In yet another embodiment, the isolated antibody of the
present
invention that binds to tau recognizes an epitope within the amino acid
sequence of
SEQ ID NO: 8 (GGKVQIINKK). Preferably, the isolated antibody recognizes an
epitope
within at least three contiguous amino acids of SEQ ID NO: 8, including within
at least 4
contiguous amino acids of SEQ ID NO: 8, within at least 5 contiguous amino
acids of
SEQ ID NO: 8, within at least 6 contiguous amino acids of SEQ ID NO: 8, within
at least
7 contiguous amino acids of SEQ ID NO: 8, within at least 8 contiguous amino
acids of
SEQ ID NO: 8, within at least 9 contiguous amino acids of SEQ ID NO: 8, and
within at
least 10 contiguous amino acids of SEQ ID NO: 8. In an exemplary embodiment,
an
isolated antibody of the present invention that recognizes an epitope within
SEQ ID NO:
8 is the antibody HJ9.3.
[0084] A preferred antibody is a humanized form of mouse antibody
derived from a hybridoma designated HJ8.5. As used herein, the term "derived
from"
means that the "derived" antibody comprises at least one CDR region from the
antibody
produced hybridoma HJ8.5. Stated another way, the "derived antibody" comprises
at
least one CDR region comprised of the amino acid sequence selected from the
group
consisting of SEQ ID NO: 16, 17, 18, 19,20 and 21.
[0085] In one embodiment, an antibody of the invention may be
derived
from the hybridoma HJ8.5, and may be encoded by a nucleic acid sequence
comprising
90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identity to the light chain
variable region of
SEQ ID NO:12, or may be encoded by a nucleic acid sequence comprising 90, 91,
92,
93, 94, 95, 96, 97, 98, or 99% identity to the heavy chain variable region of
SEQ ID
26

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
NO:13. In another embodiment, an antibody of the invention may be derived from
the
hybridoma HJ8.5, and may comprise an amino acid sequence with 90, 91, 92, 93,
94,
95, 96, 97, 98, or 99% identity to the light chain variable region of SEQ ID
NO:14, or
may comprise an amino acid sequence with 90, 91, 92, 93, 94, 95, 96, 97, 98,
or 99%
identity to the heavy chain variable region of SEQ ID NO:15. In each of the
above
embodiments, the antibody may be humanized.
[0086] In an exemplary embodiment of an antibody of the invention
that
binds to tau, the antibody comprises the light chain nucleic acid sequence of
SEQ ID
NO:12 and the heavy chain nucleic acid sequence of SEQ ID NO:13 [i.e. the
monoclonal antibody referred to herein as HJ8.5]. In another exemplary
embodiment of
an antibody of the invention that binds to tau, the antibody comprises the
light chain
amino acid sequence of SEQ ID NO:14 and the heavy chain amino acid sequence of

SEQ ID NO:15 [i.e. the monoclonal antibody referred to herein as HJ8.5].
[0087] In one embodiment, an antibody of the invention may comprise
a
light chain CDR1, such as antibody 1 of Table B. In another embodiment, an
antibody
of the invention may comprise a light chain CDR2, such as antibody 4 of Table
B. In yet
another embodiment, an antibody of the invention may comprise a light chain
CDR3,
such as antibody 6 of Table B. In an alternative embodiment, an antibody of
the
invention may comprise a combination of two or three light chain CDRs, such as
the
antibodies 2, 3, and 5 of Table B.
[0088] Similarly, in one embodiment, an antibody of the invention
may
comprise a heavy chain CDR1, such as antibody 7 of Table B. In another
embodiment,
an antibody of the invention may comprise a heavy chain CDR2, such as antibody
10 of
Table B. In yet another embodiment, an antibody of the invention may comprise
a heavy
chain CDR3, such as antibody 12 of Table B. In an alternative embodiment, an
antibody of the invention may comprise a combination of two or three heavy
chain
CDRs, such as the antibodies 8, 9, 11 of Table B.
[0089] Alternatively, an antibody of the invention may comprise one
or
more light chain CDRs and one or more heavy chain CDRs, such as the antibodies
13-
48 of Table B.
27

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Table B
Anti- Light Chain Heavy Chain
body
CDR1 CDR2 CDR3 CDR1 CDR2 CDR3
1 SEQ ID NO:16
2 SEQ ID NO:16 SEQ ID NO:17
3 SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:18
4 SEQ ID NO:17
SEQ ID NO:17 SEQ ID NO:18
6 SEQ ID NO:18
7 SEQ ID NO:19
8 SEQ ID NO:19
SEQ ID NO:20
9
SEQ ID NO:19 SEQ ID NO:20 SEQ ID NO:21
SEQ ID NO:20
11
SEQ ID NO:20 SEQ ID NO:21
12
SEQ ID NO:21
13 SEQ ID NO:16 SEQ ID NO:19
14 SEQ ID NO:16 SEQ ID NO:19
SEQ ID NO:20
SEQ ID NO:16 SEQ ID
NO:19 SEQ ID NO:20 SEQ ID NO:21
16 SEQ ID NO:16 SEQ ID NO:20
16 SEQ ID NO:16
SEQ ID NO:20 SEQ ID NO:21
17 SEQ ID NO:16
SEQ ID NO:21
19 SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:19
SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:19 SEQ
ID NO:20
21 SEQ ID NO:16 SEQ ID NO:17
SEQ ID NO:19 SEQ ID NO:20 SEQ ID NO:21
22 SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:20
23 SEQ ID NO:16 SEQ ID NO:17
SEQ ID NO:20 SEQ ID NO:21
24 SEQ ID NO:16 SEQ ID NO:17
SEQ ID NO:21
SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:18 SEQ ID NO:19
26 SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:18
SEQ ID NO:19 SEQ ID NO:20
27 SEQ ID NO:16 SEQ ID NO:17
SEQ ID NO:18 SEQ ID NO:19 SEQ ID NO:20 SEQ ID NO:21
28 SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:18
SEQ ID NO:20
29 SEQ ID NO:16 SEQ ID NO:17
SEQ ID NO:18 SEQ ID NO:20 SEQ ID NO:21
SEQ ID NO:16 SEQ ID NO:17 SEQ ID
NO:18 SEQ ID NO:21
31 SEQ ID NO:17 SEQ ID NO:19
32 SEQ ID NO:17 SEQ ID NO:19
SEQ ID NO:20
28

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
33 SEQ ID NO:17
SEQ ID NO:19 SEQ ID NO:20 SEQ ID NO:21
34 SEQ ID NO:17 SEQ ID NO:20
35 SEQ ID NO:17
SEQ ID NO:20 SEQ ID NO:21
36 SEQ ID NO:17
SEQ ID NO:21
37 SEQ ID NO:17 SEQ ID NO:18 SEQ ID NO:19
38 SEQ ID NO:17 SEQ ID NO:18 SEQ ID NO:19 SEQ ID NO:20
39 SEQ ID NO:17 SEQ ID NO:18 SEQ ID NO:19 SEQ ID NO:20 SEQ
ID NO:21
40 SEQ ID NO:17 SEQ ID NO:18 SEQ ID NO:20
41 SEQ ID NO:17 SEQ ID NO:18
SEQ ID NO:20 SEQ ID NO:21
42 SEQ ID NO:17 SEQ ID NO:18
SEQ ID NO:21
43 SEQ ID NO:18 SEQ ID NO:19
44 SEQ ID NO:18 SEQ ID NO:19 SEQ ID NO:20
45 SEQ ID NO:18 SEQ ID NO:19 SEQ ID NO:20 SEQ ID
NO:21
46 SEQ ID NO:18 SEQ ID NO:20
47 SEQ ID NO:18
SEQ ID NO:20 SEQ ID NO:21
48 SEQ ID NO:18
SEQ ID NO:21
[0090]
In various embodiments, an antibody of the invention is humanized.
For instance, in one embodiment, a humanized antibody of the invention may
comprise
a light chain variable region comprising a CDR1 of amino acid sequence SEQ ID
NO:
16 with zero to two amino acid substitutions, a CDR2 of amino acid sequence
SEQ ID
NO: 17 with zero to two amino acid substitutions, and a CDR3 of amino acid
sequence
SEQ ID NO: 18 with zero to two amino acid substitutions, or may comprise a
heavy
chain variable region comprising a CDR1 of amino acid sequence SEQ ID NO: 19
with
zero to two amino acid substitutions, a CDR2 of amino acid sequence SEQ ID NO:
20
with zero to two amino acid substitutions, and a CDR3 of amino acid sequence
SEQ ID
NO: 21 with zero to two amino acid substitutions. In a preferred embodiment, a

humanized antibody of the invention may comprise a light chain variable region

comprising a CDR1 of amino acid sequence SEQ ID NO: 16 with zero to two amino
acid
substitutions, a CDR2 of amino acid sequence SEQ ID NO: 17 with zero to two
amino
acid substitutions, a CDR3 of amino acid SEQ ID NO: 18 with zero to two amino
acid
substitutions, a heavy chain variable region comprising a CDR1 of amino acid
sequence
SEQ ID NO: 19 with zero to two amino acid substitutions, a CDR2 of amino acid
29

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
sequence SEQ ID NO: 20 with zero to two amino acid substitutions, and a CDR3
of
amino acid sequence SEQ ID NO: 21 with zero to two amino acid substitutions.
In an
exemplary embodiment, a humanized antibody of the invention may comprise a
light
chain variable region comprising a CDR1 of amino acid sequence SEQ ID NO: 16,
a
CDR2 of amino acid sequence SEQ ID NO: 17, a CDR3 of amino acid sequence SEQ
ID NO: 18, a heavy chain variable region comprising a CDR1 of amino acid
sequence
SEQ ID NO: 19, a CDR2 of amino acid sequence SEQ ID NO: 20, and a CDR3 of
amino acid sequence SEQ ID NO: 21. The invention also encompasses the
corresponding nucleic acid sequences of SEQ ID NO: 16, 17, 18, 19, 20, and 21,
which
can readily be determined by one of skill in the art, and may be incorporated
into a
vector or other large DNA molecule, such as a chromosome, in order to express
an
antibody of the invention.
II. Method of use
[0091] In an aspect, the present invention provides antibodies for
use in a
functional therapeutic composition which is administered to a living subject.
In another
aspect, the present invention provides antibodies for use in an immunoassay to
detect
tau aggregates in a sample of biological fluid obtained from a living subject.
In another
aspect, the present invention provides antibodies for use in an immunoassay to

measure the amount of tau aggregate in a sample of biological fluid obtained
from a
living subject. The amount of tau aggregate in a sample of biological fluid
obtained from
a subject can be used to classify a subject as having high or low amounts of
tau
aggregate, and may be further used to predict the risk of developing symptoms
and / or
disease associated with tau aggregation over the lifetime of the subject.
[0092] Suitable subjects include, but are not limited to, a human,
a
livestock animal, a companion animal, a lab animal, and a zoological animal.
In one
embodiment, the subject may be a rodent, e.g. a mouse, a rat, a guinea pig,
etc. In
another embodiment, the subject may be a livestock animal. Non-limiting
examples of
suitable livestock animals may include pigs, cows, horses, goats, sheep,
llamas and
alpacas. In yet another embodiment, the subject may be a companion animal. Non-

limiting examples of companion animals may include pets such as dogs, cats,
rabbits,

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
and birds. In yet another embodiment, the subject may be a zoological animal.
As used
herein, a "zoological animal" refers to an animal that may be found in a zoo.
Such
animals may include non-human primates, large cats, wolves, and bears. In
preferred
embodiments, the animal is a laboratory animal. Non-limiting examples of a
laboratory
animal may include rodents, canines, felines, and non-human primates. In
certain
embodiments, the animal is a rodent. Non-limiting examples of rodents may
include
mice, rats, guinea pigs, etc. In embodiments where the animal is a mouse, the
mouse
may be a C57BL/6 mouse, a Balb/c mouse, a 129sv, or any other laboratory
strain. In
an exemplary embodiment, the subject is a C57BL/6J mouse. In a preferred
embodiment, the subject is human.
A. Method of treatment
[0093] In an aspect, the present invention comprises a method of
reducing
the spread of tau aggregation in the brain of a subject. In another aspect the
present
invention comprises a method for reducing intracellular aggregation of tau
induced by
tau seeds. In each aspect, the method comprises administering a
pharmacologically
effective amount of anti-tau antibody to a subject. Suitable antibodies are
described
above in Section I. In a preferred embodiment, an antibody is selected from
the group
consisting of an antibody from Table 1 and an antibody from Table 2, including
a
humanized antibody, a chimeric antibody or an immunological fragment thereof.
[0094] A subject may or may not be having a symptom associated with
tau aggregation prior to administration of a pharmacologically effective
amount of anti-
tau antibody. Stated another way, a subject may or may not be experiencing a
symptom
associated with tau aggregation. A skilled artisan will appreciate that
pathological tau
aggregation likely commences prior to diagnosis or the onset of symptoms
associated
with tau aggregation. In some embodiments, a subject is having a symptom
associated
with tau aggregation. In other embodiments, a subject is not having a symptom
associated with tau aggregation. In still other embodiments, a subject has
detectable
tau pathology but is not having any other symptom associated with tau
aggregation.
Reducing the spread of tau aggregation in the brain of a subject may reduce
the
31

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
development and/or progression of symptoms associated with the pathological
aggregation of tau.
[0095] Preventing propagation of fibrillar tau aggregates may treat
pathologies associated with generation and spread of tau aggregates. As used
herein,
the terms "treating" or "treatment" include prevention, attenuation, reversal,
or
improvement in at least one symptom or sign of symptoms associated with tau
aggregation. One definition of symptoms associated with tau aggregation refers
to any
symptom caused by the formation of tau aggregates being composed of, in part,
tau
fibrils. Exemplary disorders that have symptoms associated with tau
aggregation
include, but are not limited to, progressive supranuclear palsy, dementia
pugilistica
(chronic traumatic encephalopathy), frontotemporal dementia and parkinsonism
linked
to chromosome 17, Lytico-Bodig disease (Parkinson-dementia complex of Guam),
tangle-predominant dementia, ganglioglioma and gangliocytoma,
meningioangiomatosis, subacute sclerosing panencephalitis, lead
encephalopathy,
tuberous sclerosis, Hallervorden-Spatz disease, lipofuscinosis, Pick's
disease,
corticobasal degeneration, argyrophilic grain disease (AG D), Frontotemporal
lobar
degeneration, Alzheimer's Disease, and frontotemporal dementia. Methods for
diagnosing these disorders are known in the art.
[0096] Exemplary symptoms associated with tau aggregation may
include
impaired cognitive function, altered behavior, emotional dysregulation,
seizures, and
impaired nervous system structure or function. Impaired cognitive function
includes but
is not limited to difficulties with memory, attention, concentration,
language, abstract
thought, creativity, executive function, planning, and organization. Altered
behavior
includes but is not limited to physical or verbal aggression, impulsivity,
decreased
inhibition, apathy, decreased initiation, changes in personality, abuse of
alcohol,
tobacco or drugs, and other addiction-related behaviors. Emotional
dysregulation
includes but is not limited to depression, anxiety, mania, irritability, and
emotional
incontinence. Seizures include but are not limited to generalized tonic-clonic
seizures,
complex partial seizures, and non-epileptic, psychogenic seizures. Impaired
nervous
system structure or function includes but is not limited to hydrocephalus,
Parkinsonism,
32

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
sleep disorders, psychosis, impairment of balance and coordination. This
includes
motor impairments such as monoparesis, hemiparesis, tetraparesis, ataxia,
ballismus
and tremor. This also includes sensory loss or dysfunction including
olfactory, tactile,
gustatory, visual and auditory sensation. Furthermore, this includes autonomic
nervous
system impairments such as bowel and bladder dysfunction, sexual dysfunction,
blood
pressure and temperature dysregulation. Finally, this includes hormonal
impairments
attributable to dysfunction of the hypothalamus and pituitary gland such as
deficiencies
and dysregulation of growth hormone, thyroid stimulating hormone, lutenizing
hormone,
follicle stimulating hormone, gonadotropin releasing hormone, prolactin, and
numerous
other hormones and modulators. Methods for detecting and evaluating symptoms
associated with tau aggregation are known in the art.
[0097] In some embodiments, a symptom associated with tau
aggregation
refers to dementia. Dementia is not itself a specific disease, but is an
overall term that
describes a wide range of symptoms associated with a decline in memory or
other
thinking skills severe enough to reduce a person's ability to perform everyday
activities.
Dementia is also a shared clinical feature of many diseases associated with
tau
aggregation. A skilled practitioner will be familiar with the numerous methods
available
to diagnose the severity of dementia. For example, several cognitive tests and

screening questionnaires for dementia are known in the art, all with varying
degrees of
sensitivity and specificity. Non-limiting examples include the mini mental
state
examination (MMSE), the abbreviated mental test may score (AMTS), the modified
mini
mental state exam (3MS), the cognitive abilities screening instrument (CASI),
the Trail-
making test, the clock drawing test, the Informant Questionnaire on cognitive
decline in
the elderly, the General practitioner assessment of cognition, the Clinical
Dementia
Rating (CDR), Eight-item informant interview to differentiate aging and
dementia (AD8).
[0098] In some embodiments, the severity of the symptoms of
dementia
are quantified using the CDR. Using the CDR, a score of 0 indicates no
symptoms, a
score of 0.5 indicates very mild symptoms, a score of 1 indicates mild
symptoms, a
score of 2 indicates moderate symptoms and a score of 3 indicates severe
symptoms.
Thus, any increase in a CDR score for a subject indicates a worsening in
cognition and
33

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
an increase in dementia. Moreover, change in CDR from 0 to greater than 0,
indicates
the development or onset of dementia.
[0099] In some embodiments, a symptom associated with tau
aggregation
refers to tau pathology. The term "tau pathology" refers to the pathological
aggregation
of tau. In some embodiments, tau pathology refers to neurofibrially tangles.
In other
embodiments, tau pathology refers to hyperphosphorylated tau. In still other
embodiments, tau pathology refers to a high level of tau aggregates detectable
in blood,
plasma, serum, CSF, or ISF, anywhere from 1.2 to approximately 40-fold higher
than
that detected in individuals without disease. Methods for detecting
pathological
aggregation of tau are in known in the art and further detailed in the
Examples.
[0100] In an exemplary embodiment, a method of reducing the spread
of
tau aggregation in the brain of a subject comprises administering a
pharmacologically
effective amount of anti-tau antibody to the subject, wherein the antibody is
selected
from the group consisting of an isolated antibody comprising a light chain
variable
region comprising a CDR1 of amino acid sequence SEQ ID NO: 16 with zero to two

amino acid substitutions, an isolated antibody comprising a light chain
variable region
comprising a CDR2 of amino acid sequence SEQ ID NO: 17 with zero to two amino
acid
substitutions, an isolated antibody comprising a light chain variable region
comprising a
CDR3 of amino acid sequence SEQ ID NO: 18 with zero to two amino acid
substitutions, an isolated antibody comprising a heavy chain variable region
comprising
a CDR1 of amino acid sequence SEQ ID NO: 19 with zero to two amino acid
substitutions, an isolated antibody comprising a heavy chain variable region
comprising
a CDR2 of amino acid sequence SEQ ID NO: 20 with zero to two amino acid
substitutions, and an isolated antibody comprising a heavy chain variable
region
comprising a CDR3 of amino acid sequence SEQ ID NO: 21 with zero to two amino
acid
substitutions.
[0101] In another exemplary embodiment, a method of reducing the
spread of tau aggregation in the brain of a subject comprises administering a
pharmacologically effective amount of anti-tau antibody to the subject,
wherein the
34

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
antibody specifically binds tau and recognizes an epitope comprising SEQ ID
NO: 1
(DRKDQGGYTMHQD).
[0102] In another exemplary embodiment, a method of reducing the
spread of tau aggregation in the brain of a subject comprises administering a
pharmacologically effective amount of anti-tau antibody to the subject,
wherein the
antibody specifically binds tau and recognizes an epitope consisting of SEQ ID
NO: 1
(DRKDQGGYTMHQD).
[0103] The antibodies in a pharmacologically effective amount
preferred in
pharmaceutical grade, including immunologically reactive fragments, may be
administered to a subject. Administration is performed using standard
effective
techniques, include peripherally (i.e. not by administration into the central
nervous
system) or locally to the central nervous system. Peripheral administration
includes but
is not limited to intravenous, intraperitoneal, subcutaneous, pulmonary,
transdermal,
intramuscular, intranasal, buccal, sublingual, or suppository administration.
Local
administration, including directly into the central nervous system (CNS)
includes, but is
not limited to, via a lumbar, intraventricular or intraparenchymal catheter or
using a
surgically implanted controlled release formulation.
[0104] Pharmaceutical compositions for effective administration are
deliberately designed to be appropriate for the selected mode of
administration, and
pharmaceutically acceptable excipients such as compatible dispersing agents,
buffers,
surfactants, preservatives, solubilizing agents, isotonicity agents,
stabilizing agents and
the like are used as appropriate. Remington's Pharmaceutical Sciences, Mack
Publishing Co., Easton Pa., 16Ed ISBN: 0-912734-04-3, latest edition,
incorporated
herein by reference in its entirety, provides a compendium of formulation
techniques as
are generally known to practitioners. It may be particularly useful to alter
the solubility
characteristics of the antibodies useful in this discovery, making them more
lipophilic,
for example, by encapsulating them in liposomes or by blocking polar groups.
[0105] Effective peripheral systemic delivery by intravenous or
intraperitoneal or subcutaneous injection is a preferred method of
administration to a
living patient. Suitable vehicles for such injections are straightforward. In
addition,

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
however, administration may also be effected through the mucosal membranes by
means of nasal aerosols or suppositories. Suitable formulations for such modes
of
administration are well known and typically include surfactants that
facilitate cross-
membrane transfer. Such surfactants are often derived from steroids or are
cationic
lipids, such as N-[1-(2,3-dioleoyl)propyI]-N,N,N-trimethyl ammonium chloride
(DOTMA)
or various compounds such as cholesterol hemisuccinate, phosphatidyl glycerols
and
the like.
[0106] The concentration of humanized antibody in formulations to
be
administered is an effective amount and ranges from as low as about 0.1% by
weight to
as much as about 15 or about 20% by weight and will be selected primarily
based on
fluid volumes, viscosities, and so forth, in accordance with the particular
mode of
administration selected if desired. A typical composition for injection to a
living patient
could be made up to contain from 1-5 mL sterile buffered water of phosphate
buffered
saline and about 1-5000 mg of any one of or a combination of the humanized
antibody
of the present discovery. The formulation could be sterile filtered after
making the
formulation, or otherwise made microbiologically acceptable. A typical
composition for
intravenous infusion could have volumes between 1-250 mL of fluid, such as
sterile
Ringer's solution, and 1 -1 00 mg per ml, or more in anti-tau antibody
concentration.
Therapeutic agents of the discovery can be frozen or lyophilized for storage
and
reconstituted in a suitable sterile carrier prior to use. Lyophilization and
reconstitution
may lead to varying degrees of antibody activity loss (e.g. with conventional
immune
globulins, IgM antibodies tend to have greater activity loss than IgG
antibodies).
Dosages administered are effective dosages and may have to be adjusted to
compensate. The pH of the formulations that are generally of pharmaceutical
grade
quality will be selected to balance antibody stability (chemical and physical)
and comfort
to the patient when administered. Generally, a pH between 4 and 8 is
tolerated. Doses
will vary from individual to individual based on size, weight, and other
physio-biological
characteristics of the individual receiving the successful administration.
[0107] As used herein, the term "effective amount" means an amount
of a
substance such as a compound that leads to measurable and beneficial effects
for the
36

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
patient administered the substance, i.e., significant efficacy. The effective
amount or
dose of compound administered according to this discovery will be determined
by the
circumstances surrounding the case, including the compound administered, the
route of
administration, the status of the symptoms being treated and similar patient
and
administration situation considerations among other considerations. In an
aspect, a
typical dose contains from about 0.01 mg/kg to about 100 mg/kg of an anti-tau
antibody
described herein. Doses can range from about 0.05 mg/kg to about 100 mg/kg,
more
preferably from about 0.1 mg/kg to about 50 mg/kg, or from 0.5 mg/kg to about
50
mg/kg. The frequency of dosing may be daily or once, twice, three times or
more per
week or per month, as needed as to effectively treat the symptoms.
Alternatively, the
frequency of dosing may be at least once every three months, as needed as to
effectively treat the symptoms. For example, dosing may be about every 5
weeks, about
every 6 weeks, about every 7 weeks, about every 8 weeks, about every 9 weeks,
about
every 10 weeks, about every 11 weeks, or about every 12 weeks.
[0108] The timing of administration of the treatment relative to
the disease
itself and duration of treatment will be determined by the circumstances
surrounding the
case. Treatment could begin after diagnosis of a disease associated with tau
aggregation. Alternatively, treatment could begin after clinical confirmation
of a
symptom associated with tau aggregation. Further still, treatment could begin
after
detection of tau pathology. Treatment could begin immediately in a hospital or
clinic, or
at a later time after discharge from the hospital or after being seen in an
outpatient
clinic. Duration of treatment could range from a single dose administered on a
one-time
basis to a life-long course of therapeutic treatments.
[0109] Although the foregoing methods appear the most convenient
and
most appropriate and effective for administration of proteins such as
humanized
antibodies, by suitable adaptation, other effective techniques for
administration, such as
intraventricular administration, transdermal administration and oral
administration may
be employed provided proper formulation is utilized herein.
37

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0110] In addition, it may be desirable to employ controlled
release
formulations using biodegradable films and matrices, or osmotic mini-pumps, or
delivery
systems based on dextran beads, alginate, or collagen.
[0111] Typical dosage levels can be determined and optimized using
standard clinical techniques and will be dependent on the mode of
administration.
B. Method of detecting tau aggregates in biological fluid
[0112] In an aspect, the invention provides means to detect tau
aggregate
in a sample of biological fluid obtained from a subject. In another aspect,
the invention
provides means to measure the amount of tau aggregate in a sample of
biological fluid
obtained from a subject. The method generally comprises (i) obtaining a sample
of a
biological fluid from a subject, and (ii) measuring the amount of tau
aggregate in the
sample using an antibody that specifically binds tau. Suitable antibodies are
described
above in Section I. Suitable subjects are described above.
[0113] As used herein, the term "biological fluid" refers to a
fluid obtained
from a subject. Any biological fluid comprising a tau aggregate is suitable.
Non-limiting
examples include blood, plasma, serum, urine, CSF and ISF. The fluid may be
used "as
is", the cellular components may be isolated from the fluid, or a protein
fraction may be
isolated from the fluid using standard techniques.
[0114] As will be appreciated by a skilled artisan, the method of
collecting
a sample of biological fluid can and will vary depending upon the nature of
the biological
fluid and the type of analysis to be performed. Any of a variety of methods
generally
known in the art may be utilized to collect a sample of biological fluid.
Generally
speaking, the method preferably maintains the integrity of the sample such
that tau
aggregate can be accurately detected and the amount measured according to the
invention.
[0115] Once a sample is obtained, it is processed in vitro in order
to detect
and measure the amount of tau aggregate using an anti-tau antibody. In some
embodiments, the concentration of tau aggregate in the sample is increased
prior to
detection and measurement. In some embodiments, tau aggregate is
immunoprecipitated from a sample prior to detection and measurement using at
least
38

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
one isolated anti-tau antibody. In other embodiments, tau aggregate is
immunoprecipitated from a sample prior to detection and measurement using at
least
two isolated anti-tau antibodies. In embodiments where at least two antibodies
are used
to immunoprecipitate tau aggregates, preferably a first antibody binds a first
epitope of
tau and a second antibody binds a second, non-overlapping epitope of tau. The
use of
two antibodies that bind two distinct epitopes of tau may be more efficient at
capturing
all possible tau aggregate conformers. Non-limiting examples of suitable
antibody pairs
for immunoprecipitation are listed in Table C. In a preferred embodiment, tau
aggregate
is immunoprecipitated from a sample prior to detection and measurement using
at least
two isolated anti-tau antibodies, wherein at least a first antibody recognizes
an epitope
within SEQ ID NO: 1 and at least a second antibody recognizes an epitope
within SEQ
ID NO: 8. A skilled artisan will be able to determine with routine
experimentation
whether or not tau aggregate in a sample needs to be concentrated or
immunoprecipitated prior to detection and measurement, and will be able to do
so using
methods known in the art.
Table C
Second Antibody
HJ8.1. HJ8.1. HJ8.2 HJ8.3 HJ8.4 HJ8.5 HJ8.7 HJ8.8 HJ9.1 HJ9.2 HJ9.3 HJ9.4
HJ9.5
1 2
HJ8.1.
X
1 X X X X X X X X X X
HJ8.1.
X X X X X X X X X X X
2
.., HJ8.2 X X X X X X X X X X X
=I
0
..i HJ8.3 X X X X X X X X X X X
'4 HJ8= 4 X X X X X X X X X X
X
't
z HJ8.5 X X X X X X X X X X X
HJ8.7 X X X X X X X X X X X X
HJ8.8 X X X X X X X X X X X X
HJ9.1 X X X X X X X X X X X X
HJ9.2 X X X X X X X X X X
HJ9.3 X X X X X X X X X X X X X
HJ9.4 X X X X X X X X X X
HJ9.5 X X X X X X X X X X
39

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0116] Methods for detecting and measuring an amount of protein
using
an antibody are well known in the art. All suitable methods for detecting and
measuring
an amount of protein using an antibody known to one of skill in the art are
contemplated
within the scope of the invention. Non-limiting examples include an ELISA, a
sandwich
immunoassay, a radioimmunoassay, an immunoblot or Western blot, flow
cytometry,
immunohistochemistry, and an array.
[0117] In general, an antibody-based method of detecting and
measuring
an amount of tau aggregate comprises contacting some or all of the sample
comprising
tau aggregate with an anti-tau antibody under conditions effective to allow
for formation
of a complex between the antibody and the tau aggregate. Typically, the entire
sample
is not needed, allowing one skilled in the art to repeatedly detect and
measure the
amount of tau aggregate in the sample over time. The method may occur in
solution, or
the antibody or tau aggregate may be immobilized on a solid surface. Non-
limiting
examples of suitable surfaces include microtitre plates, test tubes, beads,
resins, and
other polymers. Attachment to the substrate may occur in a wide variety of
ways, as will
be appreciated by those in the art. For example, the substrate and the
antibody may be
derivatized with chemical functional groups for subsequent attachment of the
two. For
example, the substrate may be derivatized with a chemical functional group
including,
but not limited to, amino groups, carboxyl groups, oxo groups or thiol groups.
Using
these functional groups, the antibody may be attached directly using the
functional
groups or indirectly using linkers. An anti-tau antibody may also be attached
to the
substrate non-covalently. For example, a biotinylated anti-tau antibody may be

prepared, which may bind to surfaces covalently coated with streptavidin,
resulting in
attachment. Alternatively, an antibody may be synthesized on the surface using

techniques such as photopolymerization and photolithography.
[0118] Contacting the sample with an antibody under effective
conditions
for a period of time sufficient to allow formation of a complex generally
involves adding
the anti-tau antibody composition to the sample (or to the immunopreicipitated
or
concentrated tau aggregate) and incubating the mixture for a period of time
long enough
for the anti-tau antibody to bind to any antigen present. After this time, the
complex may

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
be washed and then the complex is detected and the amount measured by any
method
well known in the art. Methods of detecting and measuring an amount of an
antibody -
polypeptide complex are generally based on the detection of a label or marker.
The
term "label", as used herein, refers to any substance attached to an antibody,
or other
substrate material, in which the substance is detectable by a detection
method. Non-
limiting examples of suitable labels include luminescent molecules,
chemiluminescent
molecules, fluorochromes, fluorescent quenching agents, colored molecules,
radioisotopes, scintillants, biotin, avidin, stretpavidin, protein A, protein
G, antibodies or
fragments thereof, polyhistidine, Ni2+, Flag tags, myc tags, heavy metals, and
enzymes
(including alkaline phosphatase, peroxidase, and luciferase). Methods of
detecting and
measuring an amount of an antibody-polypeptide complex based on the detection
of a
label or marker are well known in the art.
[0119] In a preferred embodiment, a method for measuring the amount
of
tau aggregate in a sample is an immunoassay comprising two captures antibodies
and
a detection antibody, wherein each capture antibody is an isolated anti-tau
antibody that
recognizes a tau epitope distinct from the other, and the detection antibody
is an
isolated anti-tau antibody attached to a label. The detection antibody may be
the same
antibody as one of the two capture antibodies or, alternatively, the detection
antibody
may recognize a tau epitope not recognized by either capture antibody.
Typically, the
first capture antibody and the second capture antibody are used in an amount
from
about 10:1 to about 1:10, from about 5:1 to about 1:5, from about 3:1 to about
1:3, or
from about 2:1 to about 1:2. In some embodiments, the first capture antibody
and the
second capture antibody are used at about equivalent concentrations. Non-
limiting
examples of suitable pairs of capture antibodies include the antibodies
disclosed in
Table D and Table E. Non-limiting examples of suitable detection antibodies
include the
antibodies listed in Table A, as well as antibodies that specifically bind tau
and
recognize an epitope within an amino acid sequence selected from the group
consisting
of SEQ ID NOs: 1-11. In an exemplary embodiment, a first capture antibody is
an
isolated antibody that specifically binds tau and recognizes an epitope within
SEQ ID
NO: 7, a second capture antibody is an isolated antibody that specifically
binds tau and
41

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
recognizes an epitope within SEQ ID NO: 8, and a detection antibody is an
isolated
antibody that specifically binds tau and recognizes an epitope within SEQ ID
NO: 8.
Table D First and Second Capture Antibodies
Second Capture Antibody
HJ8.1. HJ8.1. HJ8.2 HJ8.3 HJ8.4 HJ8.5 HJ8.7 HJ8.8 HJ9.1 HJ9.2 HJ9.3 HJ9.4
HJ9.5
1 2
HJ8.1.
X
1 X X X X X X X X X X
HJ8.1.
X X X X X X X X X X X
"al 2
0
-0 8¨, HJ .2 X X X X X X X X X X
X
-e! HJ8.3 X X X X
X X X X X X X
cu
= HJ8.4 X X X X X X X X X
X X
:
*4 HJ8.5 X X X X X X X X X X X
0
C..)
HJ8.7 X X X X X X X X X X X X
z
=¨, = HJ8.8 X X X X X X X X X X X
X
.lo
HJ9.1 X X X X X X X X X X X X
HJ9.2 X X X X X X X X X X
HJ9.3 X X X X X X X X X X X X X
HJ9.4 X X X X X X X X X X
HJ9.5 X X X X X X X X X X
Table E First and Second Capture Antibodies: each antibody specifically binds
tau and
recognizes an epitope within the amino acid sequence indicated by the SEQ ID
NO
shown.
Second Capture Antibody
SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID
SEQ ID SEQ ID SEQ ID
NO: 1 NO: 2 NO: 3 NO: 4 NO: 5 NO: 6
NO: 7 NO: 8
>+ SEQ ID
V X X X X X X X
0
.0
7. SEQ ID
a
ct X X X X X X X
NO: 2
= SEQ ID
0. X X X X X X X
as NO: 3
0
SEQ ID
i.c:
X X X X X X X
NO: 4
i:
SEQ ID
X X X X X X X
NO: 5
42

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
SEQ ID
X X X X X X X
NO: 6
SEQ ID
X X X X X X X
NO: 7
SEQ ID
X X X X X X X
NO: 8
[0120] In another aspect, the invention provides means to classify
a
subject based on the amount of tau aggregate measured in a sample of
biological fluid
obtained from the subject. The method generally comprises (i) obtaining a
sample of a
biological fluid from a subject and measuring the amount of tau aggregate in
the sample
using an antibody that specifically binds tau, (ii) comparing the amount of
tau aggregate
in the sample to a reference value, and (iii) classifying the subject as
having a high or
low amount of tau aggregate based on the amount of tau aggregate measured in
the
sample. Methods for obtaining a sample of a biological fluid from a subject
and
measuring the amount of tau aggregate in the sample using an antibody that
specifically
binds tau are detailed above and further described in the Examples.
[0121] Any suitable reference value known in the art may be used.
For
example, a suitable reference value may be the amount of tau aggregate in a
sample of
biological fluid obtained from a subject, or group of subjects, of the same
species that
has no clinically detectable symptom of tau aggregation. In another example, a
suitable
reference value may be the amount of tau aggregate in a biological fluid
sample
obtained from a subject, or group of subjects, of the same species that has no

detectable tau pathology. In another example, a suitable reference value may
be the
amount of tau aggregate in a biological fluid sample obtained from a subject,
or group of
subjects, of the same species that has a Clinical Dementia Rating score of
zero (CDR =
0). In another example, a suitable reference value may be the background
signal of the
assay as determined by methods known in the art. In another example, a
suitable
reference value may be a measurement of the amount of tau aggregate in a
reference
sample obtained from the same subject. The reference sample comprises the same

type of biological fluid as the test sample, and may be obtained from a
subject when the
subject had no clinically detectable symptom of tau aggregation. A skilled
artisan will
43

CA 02877397 2014-12-18
WO 2014/008404
PCT/US2013/049333
appreciate that it is not always possible or desirable to obtain a reference
sample from a
subject when the subject is otherwise healthy. For example, when monitoring
the
effectiveness of a therapy, a reference sample may be a sample obtained from a

subject before therapy began. In such an example, a subject may have tau
pathology
but may not have other symptoms of tau aggregation (e.g. dementia, declined
cognition,
etc.) or the subject may have tau pathology and one or more other symptom of
tau
aggregation. In an additional example, a suitable reference sample may be a
biological
fluid from an individual or group of individuals that has been shown not to
have tau
aggregates.
[0122]
According to the invention, a subject may be classified based on
the amount of tau aggregate measured in the sample. Classifying a subject
based on
the amount of tau aggregate measured in a sample of biological fluid obtained
from the
subject may be used to identify subjects that will develop a disease and/or
symptom
associated with tau aggregation in the subject's lifetime. Generally speaking,
a subject
may be classified as having a high or low amount of tau aggregate compared to
a
reference value, wherein a high amount of tau aggregate is an amount above the

reference value and a low amount is an amount equal to or below the reference
value.
In preferred embodiments, to classify a subject as having a high amount of tau

aggregate, the amount of tau aggregate in the sample of biological fluid
compared to
the reference value is increased at least 2-fold. For example, the amount of
tau
aggregate in the sample compared to the reference value is increased at least
2-fold, at
least 5-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least
25-fold, at least 30-
fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-
fold. When the
amount of tau aggregate in the sample of biological fluid obtained from a
subject is
increased at least 2-fold compared to a reference value, and the reference
value is a
sample of the same type of biological fluid obtained from one or more disease
free
individuals with no detectable symptom of tau aggregation (or a sample
equivalent
thereto), the subject is more likely to develop a disease and/or symptom
associated with
tau aggregation in the subject's lifetime.
44

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
DEFINITIONS
[0123] As used herein, "antibody" refers to an immunoglobulin
derived
molecule that specifically recognizes tau. An antibody of the invention may be
a full
length antibody (IgM, IgG, IgA, IgE) or may be an antibody fragment (Fab,
F(ab')2,
scFv). An antibody may be chimeric or may be humanized.
[0124] As used herein, "CDR" means "complementary determining
region."
CDRs may also be referred to as hypervariable regions.
[0125] As used herein, "light chain" is the small polypeptide
subunit of the
antibody. A typical antibody comprises two light chains and two heavy chains.
[0126] As used herein, the "heavy chain" is the large polypeptide
subunit
of the antibody. The heavy chain of an antibody contain a series of
immunoglobulin
domains, with at least one variable domain and at least one constant domain.
[0127] "Humanized", as used herein, refers to the process where
monoclonal antibodies are produced using recombinant DNA to create constructs
capable of expression in human cell culture. Any known techniques for
producing these
constructs will work for purposes of the present invention.
[0128] As used herein, "single chain variable fragments" or "scFv"
or
"scFvs", refer to fusion proteins of the variable regions of the heavy and
light chains of
immunoglobulins connected via a linker. In some embodiment, the linker is a
peptide of
about 10 to 25 amino acids.
EXAMPLES
[0129] The following examples are included to demonstrate preferred
embodiments of the invention. It should be appreciated by those of skill in
the art that
the techniques disclosed in the examples that follow represent techniques
discovered
by the inventors to function well in the practice of the invention, and thus
can be
considered to constitute preferred modes for its practice. However, those of
skill in the
art should, in light of the present disclosure, appreciate that many changes
can be

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
made in the specific embodiments which are disclosed and still obtain a like
or similar
result without departing from the spirit and scope of the invention.
Introduction to Examples 1-8
[0130] Aggregation of the microtubule associated protein tau in
neurons
and glia is associated with over 20 neurodegenerative disorders including
Alzheimer
disease (AD), progressive supranuclear palsy, and frontotemporal dementia.
Recent
evidence from human studies suggests that tau pathology does not distribute
randomly
through the brain, but instead is linked to existing networks of neuronal
connectivity.
The fibrillar tau pathology of AD progresses along known anatomical
connections,
although the mechanisms by which networks degenerate are unknown. Importantly,

recent pathological studies suggest that protein aggregates can move from one
cell to
another in human and mouse brain. Moreover, fibrillar forms of recombinant,
human
disease-associated proteins such as tau, SOD-1, a- synuclein and polygutamines
are
readily taken up from the extracellular space to trigger intracellular
misfolding. These
phenomena are reminiscent of prion propagation, for which exosomes and
tunneling
nanotubes have been proposed to mediate trans-cellular spread. It is an open
question
as to whether tau aggregates might spread protein misfolding from cell to cell
via direct
cell-cell contact or through extracellular space. Furthermore, it has not yet
been
determined whether pathological tau species can mediate true trans-cellular
propagation of aggregation, whereby an aggregate is released from a "donor"
cell,
enters a second "recipient" cell, and induces further misfolding via direct
protein-protein
contact, as opposed to more indirect mechanisms. Here it is tested whether tau
fibrils
are released directly into the extracellular space and can propagate
aggregation by this
mechanism.
Example 1. Anti-tau antibodies
[0131] Two series of anti-tau antibodies were created using
standard
techniques: the HJ8 series (mouse monoclonal antibodies against recombinant
human
tau), and the HJ9 series (mouse monoclonal antibodies against recombinant
mouse
46

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
tau) (Table 1). Binding epitopes have been mapped for many of the antibodies
(Table
A).
Table 1 HJ8 series and HJ9 series against human and
mouse tau
Antibody Isotype Application
HJ 8.1 IgG2b IgG1 IF WB IHC(h&m)
HJ 8.2 IgG2b IF WB IHC(h&m)
HJ 8.3 IgG2b IF WB IHC(h&m)
HJ 8.4 IgG1 IF WB IHC(h&m)
HJ 8.5 IgG2b IF WB IHC(h) ELISA for coating staining 3mon old
mice
HJ 8.7 IgG2b IF WB IHC(h&m) HJ8.7B for ELISA detact staining
HJ 8.8 IgG2b IF WB IHC(h&m) staining
HJ 9.1 IgG2b IF WB IHC ELISA
HJ 9.2 unknown IF WB IHC ELISA for coating staining
HJ 9.3 IgG2b IF WB IHC ELISA for coating
HJ 9.4 IgG2b IF WB IHC
HJ 9.5 IgG2b IF WB IHC
IP=Immunoprecipitation; WB= Western] Blot; ELISA = Enzyme-linked lmmunosorbent

Assay, IHC = immunohistochemistry; h=human; m=mouse
[0132] To characterize the binding affinity of the HJ8 and HJ9 series
antibodies to mouse tau and human tau, Biacore's SPR technology was used.
Biacore
sensor chip CM-5 (Carboxymethylated dextran matrix) was activated by using EDC
(1-
ethyl-3-(3-dimethylaminopropy1)-carbodiimid) and NHS (N-hydroxysuccinimide) in
1:1
ratio. 2). Then ligand, either mouse tau or human Tau, were immobilized
(2Oug/ml, in 10
mM sodium acetate pH 3.5) on Biacore CM-5 sensor chip at a flow rate of 5
I/min. The
remaining unbound area on the Biacore CM-5 sensor chip was deactivated by
passage
of 1 M ethanolamine pH 8.5.
[0133] Following preparation of the sensor chip surface, analytes (e.g.
antibody) were injected with different concentrations (0.78nM ¨ 400nM) in
filtered,
degassed 0.01 M Hepes buffer, 0.15 M NaCI, 0.005% surfactant P20, pH 7.4 at a
flow
47

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
rate of10 I/min. All the samples were run in duplicates. After each cycle/run
with single
antibody concentration, the surface of the chip was regenerated by using 10 mM
glycine
pH 1.7, to remove the bound antibody/analyte, leaving the
monomer/fibrils/ligand
attached to the surface.
[0134] From the SPR sensorgram (FIG. 2-13), the rate of association
or
On rate (Ka), the rate of dissociation or Off rate (Ka) and the affinity
constant or Affinity
of interaction (KD, where KD = Kd/Ka) were obtained (Tables 2 and 3).
Table 2 Binding data of HJ8 series and HJ9 series to mouse
tau
Antibody Ka (1/MS) Kd (us) KD (M)
HJ 8.1 3.17x 104 1.83 x 10-8 0.578 pM
HJ 8.2 2.25 x 105 3.45 x 10-7 1.57 pM
HJ 8.3 1.46 x 105 7.05 x 10-8 0.48 pM
HJ 8.4 2.78 x 105 1.03 x 10-7 0.37 pM
HJ 8.5 No binding detected
HJ 8.7 7.03 x 105 2.41 x 10-8 0.34 pM
HJ 8.8 1.92 x 105 1.78 x 10-4 0.926 nM
HJ 9.1 3.52 x 105 7.61 x 10-9 0.02 pM
HJ 9.2 2.65 x 105 1.08 x 10-4 0.4 nM
HJ 9.3 8.61 x 104 9.16 x 10-6 0.1 nM
HJ 9.4 2.28 x 105 5.1 x 10-7 2.24 pM
HJ 9.5 3.4 x 105 5.37 x 10-7 1.58 pM
Table 3 Binding data of HJ8 series and HJ9 series to human
tau
Antibody Ka (1/MS) Kd (us) KD (M)
HJ 8.1 2.43 x 104 3.19 x 10-8 1.32 pM
HJ 8.2 1.98 x 105 8.95 x 10-7 4.51 pM
HJ 8.3 1.44x 105 1.93 x 10-3 0.07 pM
HJ 8.4 2.46 x 105 3 x 10-8 0.122 pM
HJ 8.5 1.3 x 105 4.34 x 10-8 0.336 pM
HJ 8.7 6.8 x 104 2.33 x 10-8 0.34 pM
48

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
HJ 8.8 1.6 x 105 9.57 x 10-7 5.95 pM
HJ 9.1 3.1 x 105 1.84x 10-3 0.5 pM
HJ 9.2 6.13 x 104 1.15 x 10-3 24.6 nM
HJ 9.3 7.55 x 104 7.51 x 10-6 99 pM
HJ 9.4 1.53x 105 1.07x 10-3 6.9 nM
HJ 9.5 5.14x 105 1.97x 10-3 3.82 nM
Example 2. Full length tau is present in ISF.
[0135] Tau was immunoprecipitated from ISF samples of both wild-
type
mice and P3015 human tau transgenic mice (P301S tg mice, details in Methods)
using
tau antibodies recognizing both mouse and human tau. Two anti-tau monoclonal
antibodies that worked well in immunoprecipitation assays were used, as the
amount of
monomeric tau in ISF is relatively low. Following immunoprecipitation, tau was
analyzed
by immunoblot. Endogenous murine tau isoforms migrate at 48-62 kDa. In wild-
type
brain lysate, tau appeared in four separate bands on SDS-PAGE (FIG. 14A). The
most
abundant species in wild-type mice migrated at 48 kDa. In P3015 tg mice brain,
in
addition to the four endogenous murine tau bands, overexpressed human 1N4R tau

was observed as an intense band migrating at 55 kDa as well as a 39 kDa band,
which
may represent a tau degradation product.
[0136] In contrast to total brain lysates, upon immunoprecipitation
a single
tau band was detected with antibody HJ9.3 recognizing the microtubule binding
region
(MTBR) of tau in ISF from wild-type mice (FIG. 14B). This band corresponded to
the
largest isoform 2N4R observed in mouse brain lysate. In ISF of P3015 tg mice,
a
human-specific tau band was co-precipitated with the aforementioned mouse tau
band
and was slightly lower in molecular weight (FIG. 14B). These two bands were
also
precipitated by another mouse monoclonal antibody raised against tau HJ8.1
(FIG.
14C). These data suggested that the major species in ISF that is assessed by
ELISA is
likely full-length monomeric tau.
49

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Example 3. Tau RD proteins form fibrillar aggregates in transfected HEK293
cells.
[0137] The tau gene encodes six protein isoforms, and multiple
mutations
cause dominantly inherited neurodegenerative disease. Depending on splicing,
the tau
protein has either three or four repeat regions that constitute the
aggregation-prone core
of the protein, which is termed the repeat domain (RD). Expression of the tau
RD
causes pathology in transgenic mice, and there is evidence for truncation of
full-length
tau to form fragments that comprise fibrils in patients. This construct was
used rather
than full-length tau because it reliably forms fibrils in cultured cells.
Various mutations
known to increase tau aggregation were engineered into a four-repeat RD
protein:
AK280 (termed AK), P301 L, and V337M. The P301L and V337M mutants were
combined in one protein (termed LM) to create a mutant form of RD with
strongly
increased aggregation potential, similar to what has been described
previously. This
"nonphysiologic" mutant facilitates assays of transfer events and trans-
cellular
propagation of misfolding that depend on efficient formation of intracellular
aggregates,
and complements similar, but less robust aggregation phenotypes of the
"physiologic"
AK mutant. Also engineered were two proline substitutions into the AK mutant,
I227P
and 1308P (termed PP), which inhibit 13-sheet formation and fibrillization,
although they
do not block formation of amorphous aggregates. Each form of mutant tau was
fused
either at the carboxyl terminus to cyan or yellow fluorescent protein (CFP or
YFP), or to
an HA tag. Constructs are diagrammed in FIG. 15A.
[0138] To evaluate the characteristics of tau RD intracellular
aggregates,
the various forms of RD were transiently transfected into HEK293 cells. Atomic
force
microscopy (AFM) was used to evaluate SDS-insoluble material. RD(AK)-HA and
RD(LM)-HA produced evident fibrillar species (FIG. 15B). RD(AK)-HA and RD(LM)-
HA
aggregates within cells also stained positive for X- 34, a thioflavin
derivative that labels
beta sheet fibrils and emits in the blue spectrum (FIG. 15C). Additionally,
detergent
fractionation was used to test whether the inclusions visible by light
microscopy had a
biochemical correlate. In SDS insoluble pellets (1 /o Triton X-100 in 1X PBS
with
protease inhibitors for isolation of soluble pellet followed by SDS/RIPA
extraction of

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
insoluble pellets), monomer and higher molecular weight species consistent
with
oligomers were detected (FIG. 150).
[0139] The applicants previously used fluorescence resonance energy
transfer (FRET) to quantitate intracellular huntingtin protein aggregation. To
test
whether this method could be used to track tau RD aggregation, the various RD
mutants (wt, AK, PP, LM) were fused to yellow fluorescent protein (YFP: FRET
acceptor) and cyan fluorescent protein (CFP: FRET donor). These constructs
were co-
transfected into HEK293 cells (denoted as RD-CFP/RD-YFP), and intracellular
aggregate formation was quantified using FRET acceptor photobleaching confocal

microscopy and spectral emission FRET using a fluorescence plate reader (FPR).
For
confocal microscopy, cells co-expressing RD(LM)-CFP/ RD(LM) YFP were imaged
and
donor signal was measured before and after partial and complete acceptor
photobleaching. The increase in donor signal after photobleaching resulted in
a mean
FRET efficiency of 18.2% 0.058 (n=6, data are standard deviation)
confirming inter-
molecular interactions between the FRET-paired RD species (FIG. 16A). To
measure
RD-CFP/YFP aggregation by spectral FRET with a FPR, established methods were
used. This was based on co-transfection of RD-YFP and RD-CFP in a 3:1 ratio,
to
maximize donor quenching within the limits of signal detection. Significant
FRET from
RD(PP)-CFP/YFP was not observed. However, RD(AK)-CFP/YFP and RD(LM)-
CFP/YFP each produced a strong FRET signal (FIG. 16B), corroborating the
microscopy findings.
[0140] It has been previously observed that a variety of cells will
take up
recombinant tau fibrils from the extracellular media. This triggers
intracellular
fibrillization of natively folded, full length tau protein fused to YFP. To
confirm this
phenomenon, FRET was used to monitor aggregation of RD(AK)-CFP/YFP induced by
various amounts of recombinant RD fibrils. HEK293 cells were co-transfected
with
RD(AK)- CFP/YFP and cultured for 15 h. Various concentrations of RD-HA fibrils

(monomer equivalents of 0.01, 0.03, 0.1 and 0.3 pM) were then added to the
media for
9 h. Fibrils were then removed by changing the media, and the cells were
allowed to
recover for 4 h before being fixed and analyzed using FRET. A dose dependent
51

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
increase in the FRET signal induced by recombinant fibrils relative to
untreated RD(AK)-
CFP/YFP cells was observed (FIG. 16C). In summary, a correlation between
microscopic, molecular, biochemical, and biophysical measures of tau RD
aggregation
and fibril formation within cells was observed. Within certain limits,
especially with
controls for protein expression levels, the plate reader-based FRET assay
provides a
facile measure of this process.
Example 4. Trans-cellular induction of RD aggregation
[0141] The applicants have previously determined that tau
inclusions from
one cell will transfer to naïve cells in co-culture. However it has not yet
been
demonstrated that these transferred aggregates can induce further aggregation
in the
recipient cells, nor whether induction of aggregation is based on direct
protein-protein
interaction. First tested was whether RD(LM)-HA aggregates derived from one
donor
cell population would form inclusions with RD(AK)-YFP in a different recipient

population upon co-culture. One group of cells was transfected with
aggregation-prone
RD(LM)- HA, and a separate group transfected with RD(AK)-YFP. The next day,
the cell
populations were re-plated together and co-cultured for 48 h. After fixation,
they were
immunostained using an HA antibody, and counterstained with X-34. Many cells
were
observed with RD(LM)-HA and RD(AK)-YFP co-localized in inclusions (FIG. 17A).
Frequently these inclusions also stained positive for X-34, indicating beta
sheet
structure. These studies were extended by using the FRET assay to monitor
aggregation of RD(AK)-CFP/YFP induced by co-culture with cells expressing
RD(LM)-
HA. In this case, two populations of cells were co-cultured. The donor
population
expressed RD(LM)-HA and the recipient population expressed RD(AK)- CFP/YFP.
The
[3- sheet-resistant form of tau RD(PP)-HA or mock transfected cells were used
as
negative controls. After 48 h FRET was measured from the cell monolayers. A
strong
increase in FRET induced by co-culture with RD(LM)-HA versus RD(PP)-HA or mock

transfected cells was observed (FIG. 17B). A small increase in FRET signal was

observed following co-culture of RD(LM)-HA cells with RD(WT)-CFP/YFP recipient
cells
(data not shown). These results suggested movement of one aggregation-prone
tau
52

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
species from one cell to another to trigger co-localization in a beta-sheet
rich inclusion.
Aggregate release could potentially occur after cell death, however, no
evidence for this
was observed using propidium iodide staining of the various transfected
populations
(FIG. 17C).
Example 5. Propagation of misfolding by direct protein contact
[0142] While strongly suggestive, these results could not formally
address
whether co-aggregation occurred via direct protein contact, with
intermolecular
association between tau RD derived from donor contacting the corresponding
protein in
recipient cells. FRET was used to address this question. First, RD(LM)-CFP was
co-
expressed within a donor cell population, and RD(LM)-YFP in a second recipient

population. FRET from the cell monolayers was measured after 48 with both
confocal
microscopy and the FPR. Using confocal microscopy, CFP signal was measured
before
and after photobleaching of YFP. A mean FRET efficiency of -14.2% was
recorded,
indicating that inclusions contained RD(LM)-CFP and RD(LM)-YFP in direct
contact
(FIG. 18A). Relative FRET signals were then compared via FPR, using different
forms
of unlabeled RD to induce aggregation of RD-CFP. First, RD(AK)-CFP and RD(LM)-
HA
were co-expressed within a donor cell population, and RD(AK)-YFP in a second
recipient cell population. RD(LM)-HA serves as an enhancer of both RD(AK)-CFP
aggregation and movement, prompting its subsequent transfer into the RD(AK) -
YFP
recipient cells. This led to a small but reproducible FRET signal increase in
the co-
cultured cells. This signal disappeared when either the CFP- or YFP-tagged RD
constructs contained the PP mutation that blocks 13-sheet formation (FIG.
18B),
indicating that both members of the pair must have the capacity to form a beta
sheet
structure. Taken together with the prior experiments, these results suggested
that
propagation of misfolding by direct contact occurs, i.e. an aggregate from one
cell exits
to contact and trigger misfolding of natively folded protein in a second cell.
This data
implied that amplification of misfolding might also occur in serial cell co-
cultures. It was
predicted that pre-exposure of a "donor" cell population to aggregation seeds
would
increase final aggregation detected in a recipient cell population. This was
tested by
53

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
successively culturing three populations of cells. The first population
expressed various
forms of non-fluorescent RD-HA to form aggregation "seeds." The second group
expressed CFP or RD(AK)-CFP, to be either non-permissive (CFP) or permissive
RD(AK)-CFP) for aggregate maintenance. These two groups were co-cultured for
48 h
to allow amplification of misfolding. Next, 50% of the combined first and
second groups
were then co-cultured for 48 h with a third group of cells expressing RD(AK)-
YFP. This
third recipient group served as a "reporter" to indicate the degree of RD(AK)-
CFP
intracellular aggregation and propagation. Prior exposure of RD(LM)-HA to the
RD(AK)-
CFP population increased final FRET by 2.6 fold vs. cells that had not been
pre-
exposed to aggregation-prone tau. As expected, interposition of cells
expressing pure
CFP in the second population of cells completely blocked the effect of prior
exposure to
tau RD "seeds" (FIG. 18C). Taken together these data indicate an amplification
of tau
aggregation within serially cultured cell populations.
Example 6. Cell-cell propagation mediated by release of aggregates into the
extracellular space
[0143] The mechanism by which protein aggregates move between cells
is
unknown. For example, some have postulated prion protein propagation via
tunneling
nanotubes, while others have suggested exosomes. Since antibodies against tau
protein have previously been reported to reduce pathology in vivo, it was
hypothesized
that tau aggregates might be released directly into the extracellular space.
Whereas
trans-cellular movement based on cell-cell contact should be independent of
the volume
of extracellular media, it was predicted that transcellular movement of tau
might be
sensitive to extracellular volume, as has been described for SOD1. To start,
the effect of
co-culture in the setting of various volumes of media was first tested. It was
observed
that increasing the cell culture medium volume reduced the efficiency of
transcellular
movement of aggregates (FIG. 19A). Further, transfer of conditioned medium
from cells
expressing RD(LM)-HA was sufficient to induce aggregation in cells expressing
RD-
CFP/YFP (FIG. 19B). These results were consistent with the movement of tau
between
54

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
cells through the extracellular space, but could not determine whether the
protein was
encapsulated in an endosome.
[0144] It was reasoned that access to encapsulated tau would be
blocked
by the lipid membrane, whereas free tau would be accessible to an antibody.
Thus, it
was tested whether a mouse monoclonal antibody (HJ9.3) that can
immunprecipitate
tau would block transcellular propagation. A modification of the cellular
model of tau RD
propagation described above was used, in which RD(LM)-HA and RD(AK)-CFP were
co-expressed within one cell population, and co-cultured for 48 h with cells
that express
RD(AK)-YFP, prior to analysis by FRET. HJ9.3 versus pooled mouse IgG was
tested for
the 48 h co-culture period. A dose dependent reduction in trans-cellular
propagation
with HJ9.3 was observed, while non-specific IgG had no effect (FIG. 19C and
D).
Importantly, HJ9.3 had no effect on intracellular aggregation of RD(AK)-CFP
and
RD(AK)-YFP when the two proteins were co-expressed within the same cell (FIG.
19E),
indicating the antibody was not directly inhibiting intracellular aggregation.
The role of
free tau was further tested in transcellular propagation by evaluating
induction of tau
misfolding using biochemistry. The induction of aggregation by detergent
fractionation
and Western blot was confirmed, which revealed an increase in RD(AK)-YFP in
the
insoluble fraction induced by co-culture with RD(LM)-HA. HJ9.3 blocked the
effect of
RD(LM)-HA to induce insolubility of RD-YFP in co-cultured cells (FIG. 19F and
G).
[0145] The effectiveness of antibody addition suggested that free
tau was
directly transferring between cells, but left uncertain the mechanism of
antibody
inhibition. It was hypothesized that HJ9.3 was blocking uptake of tau fibrils
into cells. To
test this idea flow cytometry was used to monitor the effect of the antibody
on trans-
cellular movement of aggregates. The applicants have previously established a
cytometry paradigm whereby one population of cells is labeled with mCherry,
and the
second contains tau-YFP fusions. After co-culture, it is possible to monitor
trans-cellular
movement based on the relative percentage of dual-positive (YFP/mCherry)
cells. A
population of HEK293 cells was transfected with tau RD(LM)-YFP, and a second
population was transduced with lentivirus expressing mCherry. After washing
and
resuspending the two populations, the cells were then co-cultured for 48 h in
the

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
presence or absence of 10-fold dilutions of HJ9.3 in the medium. Cells were
harvested
and the relative number of dual positive cells measured using flow cytometry.
Negative
controls consisted of the same cell populations mixed prior to sorting. Each
data point
consisted of biological triplicates. Co-cultured cells had significantly more
RD(LM)-
YFP/mCherry dual positive cells (2.07%) compared to 0.142% of premixed cells
(background). HJ9.3 decreased the percentage of dual positive cells from 2.07%
to
1.31% (FIG. 19H). This parallels the effect of this antibody on transcellular
propagation
of aggregation as measured by FRET. The difference in the potency of this
antibody in
blocking propagation as measured by FRET and flow cytometry is most probably
due to
the differences between the two techniques used to measure this event.
[0146] To further monitor the effect of the HJ9.3 antibody on trans-
cellular
movement of aggregates, direct immunofluorescence was used in an attempt to
define
where the HJ9.3/antibody complexes deposited. RD(AK)-YFP cells or non-
transfected
cells were cultured in the presence of HJ9.3 for 48 hrs. Cells were fixed with
4% PFA,
permeabilized with 0.25% TritonX-100 and then exposed to goat anti-mouse Alexa
546
labeled secondary antibody. A very small number of HJ9.3/tau complexes were
present
inside cells. However, most complexes were found outside of the cells, mainly
bound to
the cell membrane. This antibody decoration was not present in nontransfected
cells
indicating that the signal is specific to the HJ9.3/tau complexes (FIG. 20).
Thus HJ9.3
blocks tau aggregate uptake, trapping aggregates outside the cell.
Example 7. Tau fibrils mediate cell-cell propagation
[0147] The activity of HJ9.3 in the propagation assay created an
opportunity to define the tau species responsible. HJ9.3 was used to extract
tau from
the cell media. HJ9.3 or control IgG was added to the media of cells
expressing a
variety of RD constructs (wt, PP, AK, LM). Antibodies were added either at the

beginning or the end of the 48 h culture period. Media were harvested for
affinity
purification of antibody/antigen complexes using protein-G-agarose beads. The
complexes were washed, and then boiled in SDS loading buffer for analysis by
Western
blot. HJ9.3 specifically captured tau RD species from the cell media, while
IgG had no
56

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
appreciable effect (FIG. 21A). A -10-fold increase in the tau protein present
in the
media was observed when HJ9.3 was present throughout the culture period, as
opposed to addition at the end of this period (FIG. 21B). Higher-order
molecular weight
species were also noted in the media of RD(AK)-HA and RD(LM)-HA transfected
cells,
consistent with RD aggregates. RD(PP)-HA tau had the least protein present in
the
medium, and no higher-order species were observed on Western blot. A time
course
(Oh, 3h, 6h, 9h, 12h, 24h and 48 h) of the previously described experiment
showed a
time-dependent increase in the levels of tau in the media, implying that HJ9.3
incubation
was indeed increasing the steady-state level of tau protein present in the
conditioned
medium (FIG. 21C). Taken together, these data indicated that HJ9.3 blocks cell-
to-cell
propagation by interference with aggregate uptake into cells, and is
consistent with a
steady state flux of tau aggregates in and out of cells.
[0148] The precise nature of the tau species that mediate trans-
cellular
propagation is not known. Thus, HJ9.3 was used to trap these species for
imaging via
AFM. HEK293 cells that were transfected with the various tau mutants were
cultured in
the presence of HJ9.3. After 48 the antibody/antigen complexes were purified
with
protein-G agarose beads. The complexes were then eluted from the beads in high
salt
buffer, and deposited on AFM chips for imaging. Evident fibrillar species were
detected
in the media of cells expressing RD(AK)-HA and RD(LM)-HA, while RD(PP)-HA
produced only amorphous aggregates, (FIG. 210), and mock-transfected cells
produced no signal (data not shown). These findings are consistent with free
tau fibrils
mediating trans-cellular propagation of tau aggregation by their release into
the
extracellular space.
Example 8. Effect of anti-tau antibodies on tau pathology in vivo
[0149] The activity of two additional antibodies against full
length,
recombinant human tau were tested in the propagation assay. RD(LM)-CFP and
RD(AK)-YFP cells were co-cultured for 48 hrs in the presence and absence of
different
monoclonal antibodies that target different tau epitopes (HJ8.5, HJ9.3 and
HJ9.4, FIG.
22A). HJ3.4 antibody against Ar3 peptide was used as a negative control. All
three anti-
57

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
tau antibodies blocked the trans-cellular propagation of pro-aggregation
mutants of RD-
tau between cells (FIG. 22B). The negative control, HJ3.4, did not block trans-
cellular
propagation. HJ8.5, HJ9.3 and HJ9.4 also detected RD-tau fibrils by ELISA
(FIG. 22C).
[0150] To block the propagation of tau aggregates from cell to cell
in vivo,
a passive vaccination approach was used with antibodies targeting different
epitopes on
tau. Anti-tau antibodies, HJ8.5 and HJ9.3, or vehicle were each infused into
the lateral
ventricle of 6 month old, P301S tg mice by intracerebroventricular injection
using Alzet
osmotic pumps (2006 model, Fig 23A). Brain cannula attached to an Alzet pump
assembly were surgically implanted into the left lateral ventricle of each
mouse at the
position 0.4 mm anteroposterior to bregma, 1.0 mm lateral to midline and 2.5
mm
dorsoventral (Fig 23B). After treatment, placement of the cannula was verified
by cresyl
violet staining (FIG. 23C). The Alzet osmotic pumped was replaced after 6
weeks, and
the experiment concluded on day 84.
[0151] To confirm that the experimental design did not result in
antibody
degradation and/or inactivity, antibodies were collected from the Alzet pump
after 6
weeks of infusion into mouse brain and loaded onto an SDS-PAGE gel. The gel
was
first stained by Coomassie blue dye (FIG. 24A) and then analyzed by western
blotting
using antibodies taken from the pump before and after the 6 week infusion
(FIG. 24B).
All the antibodies were stable and active after 6 weeks in the Alzet pump at
physiological temperature in vivo. It was further confirmed that spiking of
recombinant
human tau protein with different infusion antibodies did not interfere with
HJ8.7 ¨ BT2B
ELISA assay for measuring total tau (FIG. 25).
[0152] To determine whether antibody treatment reduced pathological
tau
staining, tau staining was assessed in tissue sections of the 9-month old,
P301S tg mice
treated with Vehicle/PBS or the anti-tau monoclonal antibodies. Coronal
sections of the
piriform cortex were stained with biotinylated AT8 antibody, which recognizes
an
abnormally phosphorylated form of tau. Quantitative analyses of preliminary
immunohistochemistry data showed that abnormally phosphorylated tau load was
remarkably reduced after infusion of HJ8.5 and HJ9.3 in mouse brain (FIG. 26
and 27).
Biochemical analysis of these effects are underway. If successful, passive
immunization
58

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
against tau propagation and pathology could become a therapeutic approach to
treat
Alzheimer's Disease, fronto-termporal dementia or other tauopathies.
Discussion for Examples 3-7
[0153] It has been previously proposed that prion-like mechanisms
involving templated conformational change and trans-cellular propagation of
aggregation could explain the relentless progression of tauopathies and other
neurodegenerative diseases. This would consist of the release of a protein
aggregate
from a donor cell, entry into a recipient cell, and direct contact with
natively folded
protein to amplify the misfolded state. However, mechanistic evidence to
support this
model of tauopathy has been incomplete, and trans-cellular propagation of tau
misfolding in this manner has not previously been demonstrated. Examples 3-7
now
describe transcellular propagation of tau aggregation in cultured cells via
secreted tau
aggregates, and propose a likely mechanism. First documented was spontaneous
formation of RD tau fibrils in transfected cells using X-34 staining and AFM
of extracted
material. Then observed was the coincidence of tau derived from two separate
cells in
intracellular inclusions using confocal microscopy. This was associated with
increased
detergent insolubility of tau RD(AK)-YFP upon co-culture with cells expressing
an
aggregation-prone form of the protein, RD(LM)-HA. Also documented was this
increase
in aggregation using FRET between RD(LM)-CFP/YFP that were co-expressed within

the same cells. This was detected by acceptor photobleaching (microscopy), and

spectral methods (FPR). Next used was FRET between RD(LM)-CFP and RD(LM)-YFP
expressed in separate cell populations to document that propagation occurred
by direct
protein contact. This method was then extended to document amplification of
tau
protein misfolding within the cell populations in successive culture
conditions.
Transcellular propagation of tau aggregation is mediated by fibrils that are
released
directly into the extracellular space, because transfer is sensitive to
extracellular
volume, conditioned medium can increase intracellular aggregation, and an anti-
tau
antibody (HJ9.3) interfered with cellcell propagation, and trapped
extracellular tau fibrils.
Using a variety of techniques, the applicants have thus documented the trans-
cellular
59

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
aggregate propagation via ternplated conformational change and propose a
simple
model to explain these phenomena (FIG. 29).
[0154] Trans-cellular propagation - Although spontaneous movement
of
aggregated tau between cells has been previously described, it was unknown
whether
tau protein aggregates could propagate a misfolded state between cells by
direct
contact of the proteins, as opposed to indirect effects on the cell. Cell
culture studies of
a-synuclein have also suggested propagation, but it is unclear what is the
nature of the
species (e.g. aggregates vs. dimers vs. monomer) derived from donor cells and
those
formed in recipient cells. Likewise, SOD1 aggregates can transfer between
cells via the
medium to induce further aggregation, but the precise nature of the
responsible protein
conformers, and whether direct protein-protein contact occurs is unclear.
Injection of
purified A1342 and tau fibrils into transgenic mouse brain induces aggregation
of
endogenous tau, with nearby development of tau fibrils, but it is difficult to
rule out
seeding by injected protein. Work from the Applicants' lab, and subsequently
from
others has documented movement of tau aggregates and induction of aggregation
by
recombinant protein from the outside to the inside of the cell. But no prior
study of the
tau protein has demonstrated bona fide propagation: aggregate movement from
one cell
to another, direct contact with the native protein, conversion of the protein
in the
recipient cell to a fibrillar state, and amplification of the misfolded
species.
[0155] This work demonstrated these phenomena in several ways.
First, it
was found that co-culture of an aggregation-prone form of tau RD(LM)-HA with
cells
expressing RD(AK)-YFP leads to co-localization in 13-sheet positive
inclusions. Next, it
was observed that co-culture of cells expressing RD(LM)-HA with another
population
expressing both RD(AK)-CFP and RD(AK)-YFP led to an increase of FRET signal,
suggesting that movement of RD(LM)-HA into cells expressing the FRET pair was
inducing their aggregation. To demonstrate direct contact and coaggregation of
tau
aggregates moving between cells, RD(AK)-CFP and RD(AK)- YFP were expressed in
separate populations. This led to a FRET signal derived from trans-cellular
movement
and co-aggregation that disappeared if either one of the constructs contained
a double
proline mutation to block 13-sheet formation. Induction of full-length tau-YFP
aggregation

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
by transfer of RD-CFP aggregates was also observed, but the efficiency is
reduced
(data not shown). Finally, the efficiency of FRET induced by trans-cellular
movement of
protein aggregates increased significantly by preliminary co-culture of RD(LM)-
HA
expressing cells with those expressing RD(AK)-CFP, demonstrating that an
aggregated
state can be amplified within a population of cells.
[0156] Antibody modulation of tau aggregate propagation -
Antibodies
against A6 peptide, which is predominantly extracellular, can prevent A6
aggregation in
the brain and remove existing aggregates. While there are potential side
effects, such
antibodies hold promise as treatments. However, the success of vaccination in
mouse
models of tauopathy and synucleinopathy has been puzzling in light of the fact
that the
target proteins are predominantly intracellular. It was observed that HJ9.3, a
mouse
monoclonal antibody against tau- RD, inhibited the trans-cellular propagation
of tau
aggregation. However, this antibody had no effect on intracellular aggregation
of tau.
Chronic exposure of the cell medium to this antibody strongly increased the
steady state
tau levels in the media. This was corroborated by flow cytometry studies which
indicated
that HJ9.3 blocks transfer of aggregates from one cell to another. Finally,
HJ9.3/tau
complexes trapped at the cell surface were observed. The effect of this
antibody
suggested strongly that tau fibrils are released into the extracellular space,
and are not
propagating misfolding primarily via cell-cell transfer in exosomes or
tunneling
nanotubes, as has been proposed for prions. Further, aggregates present
outside the
cell, if not trapped by HJ9.3, are likely taken up again into cells. Multiple
modes of
inhibition are conceivable for therapeutic antibodies, including
disaggregation of protein
fibrils, blockade of conversion within cells, and promotion of intracellular
degradation.
Our results with HJ9.3 are most consistent with interference with cell uptake
as one
mechanism that could be used to block tauopathy, and suggest new ways to
consider
development and optimization of therapeutic antibodies for neurodegenerative
diseases.
[0157] Trans-cellular propagation via fibrillar tau - The
effectiveness of
HJ9.3 in blocking propagation of tau aggregation allowed use of this antibody
to trap the
responsible species. lmmuno-affinity purification of tau from conditioned
medium
61

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
revealed fibrillar tau. No tau fibrils in medium from control cells were
observed, or from
those expressing the 8-sheet-resistant RD(PP)-HA, which produced amorphous
aggregates. RD(AK)- HA and RD(LM)-HA expression each caused fibril secretion
into
the extracellular space. It has been unclear how protein aggregation in one
cell might
influence the aggregation in a neighboring cell, and it was formally possible
that
cytokines, exosomes, or direct connections between cells might facilitate this
process.
These possibilities cannot be completely excluded. However, these results are
most
consistent with free fibrillar species as mediators of propagation through the

extracellular space. This work suggests answers to several important questions
about
the mechanisms by which protein aggregates propagate from one cell to another
in
culture, and thus how they might do so in vivo. In conjunction with the
methods
described here to monitor trans-cellular propagation, it may be possible to
target this
process with pharmacological and biological agents for more effective
treatment of
tauopathies and other neurodegenerative diseases.
Methods for Examples 1-8.
Antibodies
[0158] The longest mouse recombinant tau isoform mTau40 (432 aa)
and
the longest human tau isoform hTau40 (441 aa) were produced in the laboratory
of Eva
Mandelkow and used as standards in the tau ELISA. The mouse monoclonal
antibody
Tau-5, which recognizes both human and mouse tau (epitope at residues 218¨
225),
was from the laboratory of L. Binder (LoPresti et al., 1995; Porzig et al.,
2007).
Monoclonal antibodies HJ8.1 and HJ9.3 are mouse monoclonal antibodies raised
by
immunizing against human tau and mouse tau, respectively, in tau knock-out
mice (The
Jackson Laboratory). Both antibodies recognize mouse and human tau on Western
blots, by immunoprecipitation, and in ELISA assays. HJ9.3 recognizes the
microtubule
binding region (MTBR) of tau. Mouse monoclonal antibody BT-2, which also
recognizes
human and mouse tau (epitope at residues 194¨ 198), was obtained from Pierce.
Rabbit polyclonal antibody directed against Tau (ab64193, epitope located in
the repeat
domain region) was purchased from Abcam, Cambridge, MA. Mouse monoclonal
62

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
antibody directed against hemagglutinin HA (HA.11 Clone 16B12) was purchased
from
Covance, Emeryville, CA. Rabbit polyclonal GFP antibody (sc-8334) was
purchased
from Santa Cruz Biotechnology.
Plasmids
[0159] Sequences encoding the four repeat domain (RD) of the
microtubule associated protein tau were used for protein expression. In
addition to the
wild-type form, various tau mutants were created: AK280 A(K); P301L/V337M
(LM);
K280/1227P/I308P (PP). These sequences were either subcloned into pcDNA3.1
(Invitrogen) with a C-terminal hemagglutinin (HA) tag, or into pEYFP-N1 or
pECFP-N1
(Clontech) to create Cterminal fluorescent protein fusions.
Animals
[0160] P3015 tg mice (line P519), which overexpress P3015 human T34
isoform tau (1N4R), have been generated and characterized previously and are
on a
B6C3 background. P3015 tg mice were obtained from the Jackson Laboratory. Tau
knock-out mice were obtained from The Jackson Laboratory. Age and genetic
background matched nontransgenic mice littermates were used as wildtype mice.
In all
experiments, both male and female were used in this study.
lmmunoprecipitation and immunoblot analysis
[0161] lmmunoprecipitation and immunoblot analysis. Hippocampal
microdialysis samples were collected at 1.0 l/min for 15 h from P3015 tau
transgenic
mouse and wild-type mice. ISF was immunoprecipitated by Dynabeads (Invitrogen)

coated with HJ8.1 or HJ9.3 tau antibody according to the manufacturer's
instructions.
Precipitated fractions were loaded on a reducing 4-12% Bis-Tris mini-gel
(Invitrogen)
and transferred to nitrocellulose membrane. Biotinylated BT-2 antibody
(Pierce) and
Poly- HRP-conjugated streptavidin (Thermo Scientific) were used to eliminate
the
interference of precipitated antibodies. HEK293 cells were cultured in
Dulbecco's
Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum, 100
63

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
pg/mL penicillin and 100 pg/mL streptomycin. Cultures were maintained in a
humidified
atmosphere of 5% CO2 at 37 C. For transient transfections, cells plated in
Optimem
medium were transfected using Lipofectamine/Plus reagent and 600 ng of
appropriate
DNA constructs (Invitrogen, Carlsbad, CA, USA) according to manufacturer's
recommendations, and harvested 24 h or 48 h later for further analyses.
Detergent fractionation and Western blot analyses
[0162] HEK293 cells were plated at 400,000 cells/well in a 12-well
plate.
The following day cells were transfected with 600 ng of plasmid. After 48 h,
cells were
harvested with 0.05% trypsin for 3 minutes at 372C, pelleted briefly at 7000 x
g and
lysed in 100p1 of 1% Triton in PBS containing protease inhibitors. Soluble
cytosolic
proteins were then collected by centrifugation at 14,000 x g for 10 minutes.
Insoluble
proteins were obtained by resuspending the pellet in RIPA/SDS buffer and
centrifugation at 20,000 x g for 15 minutes following benzonase nuclease
digestion of
nucleic acids. For co-culture experiments, equal numbers of cells transfected
with
RD(LM)-HA and RD(AK)-YFP were co-cultured together for 48 h before harvesting
and
Western blotting. Equivalent amounts of HEK293 cell protein extract from each
fraction
were analyzed using 4%-20% polyacrylamide gels (Biorad); antibody directed
against
tau RD (which recognizes an epitope in the RD region) at a 1:2000 dilution
(ab64193,
Abcam, Cambridge, MA) and/or antibody directed against OFF at 1:1000 dilution
(sc-
8334, Santa Cruz Biotechnology, Inc.). A chemiluminescence-based peroxidase-
conjugated secondary antibody reaction was performed and detected by X-ray
film.
Quantification was performed using Image J analysis software.
Co-culture experiments: Measuring RD-CFP/YFP co-aggregation by FRET
[0163] HEK293 cells were plated at 300,000 cells/well in a 12-well
plate.
The following day, cells were transfected with 600 ng of plasmid as described
above.
Co-transfected cells received a combination of 150 ng of RD-CFP constructs and
450
ng of RD-YFP constructs. 15 h later, cells were harvested with 0.05% trypsin
for 3
minutes at 372C, and a fraction of cells were re-plated in a 96-well plate in
64

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
quadruplicate, or on ibidi p-slides (ibidi GmbH, Germany) for imaging by
microscopy.
Cells were then cultured an additional 48 h before fixation with 4%
paraformaldehyde
and analysis.
Co-culture experiments: Measuring induction of RD- YFP aggregation by RD-HA
[0164] HEK293 cells were transfected with either RD(AK)-YFP or
RD(LM)-
HA in 12-well plates. After 15 h the cells were replated together onto ibidi p-
slides and
cocultured an additional 48 h. They were then fixed and stained with anti-HA
antibody
and X-34 for analysis by microscopy.
Co-culture experiments: Propagation assays in co-culture
[0165] Two populations of HEK293 cells in a 12-well plate were co-
transfected with 300 ng RD(LM)-HA and 300 ng RD(AK) -CFP together, or with
RDA(K)
- YFP. After 15 h, equal percentages of the two populations were co-cultured
for 48 h in
a 96-well plate format. Cells were then fixed with 4% paraformaldehyde and
FRET
analysis was performed using the Fluorescent Plate Reader (FPR). For FRET
microscopy analysis, two populations of HEK293 cells in a 12-well plate were
transfected with 600ng RD(LM)-CFP or with RD(LM)-YFP. After 15 h, equal
percentages of the two populations were co-cultured for 48 h on ibidi p-
slides. Cells
were then fixed with 4% paraformaldehyde and FRET acceptor photobleaching was
conducted.
Co-culture experiments: Amplification of tau aggregation in serial culture
[0166] HEK293 cells were transfected in a 12- well plate with 600
ng of
various forms of nonfluorescent RD-HA and cultured for 24 h. A second group of
cells
were transfected with CFP or RD(AK) -CFP. Equal percentages of the first and
second
populations were then co-cultured for 48 h. At this point, 50% of this
population was
plated with a population of cells transfected with RD(AK)-YFP in a 96-well
plate for 48 h.
Cells were then fixed with 4% paraformaldehyde for FRET analyses using the FP
R.

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Media transfer and conditioned media experiments
[0167] HEK293 cells were transfected in a 12-well plate with either
600 ng
of RD(LM)-HA or co-transfected with a combination of 150 ng of RD(AK)-CFP
construct
and 450 ng of RD(AK)- YFP construct. 15 h later, cells were harvested with
0.05%
trypsin for 3 minutes at 372C. An equivalent number of cells expressing RD(AK)-

YFP/CFP and RD(LM)-HA were co-cultured for 48 h in varying amounts of cell
culture
medium. Cells were then fixed with 4% paraformaldehyde and FRET analysis was
performed. For the conditioned media experiments, 15 h after transfection,
media from
RD(LM)-HA cells containing transfection complexes was replaced with fresh
media.
Cells expressing RD(AK)- YFP/CFP were harvested with 0.05% trypsin for 3
minutes at
372C and replated in 96-well plate. 24 h later, conditioned media from cells
transfected
with RD(LM)-HA was collected and added to cells expressing RD(AK)-YFP/CFP. 48
h
later cells were fixed with 4% paraformaldehyde and FRET analysis was
performed.
Fluorescence Resonance Energy Transfer (FRET) assays: FRET measurements by
microscopy with photobleaching
[0168] HEK293 cells transfected for cotransfection and co-culture
experiments as described earlier were prepared for FRET acceptor
photobleaching
microscopy. All images were obtained using a C-Apochromat 40x 1.2 NA lens
(Carl
Zeiss Advanced Imaging Microscopy, 07740 Jena, Germany 100 X (CFP). Digital
images were acquired using a Zeiss LSM510 Meta NLO Multiphoton/Confocal laser
scanning microscope system on the Zeiss Axiovert 200M. Channels used for
imaging
were as follows: the donor CFP was stimulated using a 458nm argon laser and
fluorescence collected with a 480-520nm bandpass filter; the acceptor YFP was
stimulated using a 514nm argon laser and fluorescence collected with a long-
pass
560nm filter. To create an image in which the intensity reflected an estimate
of FRET
efficiency, the value of the initial CFP image was subtracted from the final
CFP image
obtained after photobleaching on a pixel-by-pixel basis, and this difference
was
multiplied by 100 and divided by the final CFP image intensity: 100 X (CFP
= final -
66

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
CFPinitial)/CFPfinal= Proper adjustments were made for partial acceptor
photobleaching.
Image arithmetic and grayscaleto- color image conversion were done using NIH
ImageJ
1.44 software.
FRET assays: Fluorescence Plate Reader
[0169] Spectral FRET measurements (FRET/donor) were obtained using
a
TecanM1000 fluorescence plate reader according to methods previously
described.
When donor and acceptor are not fused to the same protein, spectral FRET
measurements depend on careful control for the relative amount of donor and
acceptor
proteins expressed within the cell. All values on the plate reader were first
background
subtracted against mock-transfected cells. The YFP signal in each well
(Smp1485ex/528em FRET) was used to estimate RD-YFP expression levels, and it
was
likewise assumed that under experimental conditions that RD-CFP/YFP do not
vary
independently. This helps eliminate the possibility that changes in apparent
FRET are
due simply to variations in RD expression levels. Relative contribution of
acceptor
activation (528nm) by donor excitation signal (435nm) to the overall FRET
measurement was corrected by determining the "crossover activation" fraction
for
acceptor, X, where X=RD-YFP signal measured at 435ex/528em divided by the
signal
measured at 485ex/528em. This "crossover activation" is essentially constant
across
different expression levels of RD-YFP encountered in the experiments. The
"measured"
FRET value in each sample is recorded at 435ex/528em, the "donor" value (CFP)
is
recorded at 435ex/485em. The "actual" FRET/donor value for each well is then
reflected
as:
FRETactual = (Sr11131435ex/528em -X*(SMP1435ex/528em))/SMPI435ex/528em
[0170] This method of measuring protein aggregation by FRET has
reliably allowed detection of subtle changes in response to pharmacologic as
well as
genetic manipulations of androgen receptor and huntingtin protein aggregation
that
were corroborated by visual and biochemical analyses. Since the relative
amount of
spectral FRET measured depends on the ratio of acceptor:donor, a constant
ratio of 3:1
67

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
was used when RD-CFP and RD-YFP are co-expressed within the same cell. This
provides close to maximal FRET efficiency while allowing for acceptable signal
:noise in
the measurement of donor signal.
Atomic Force Microscopy (AFM)
[0171] RIPA-insoluble proteins were extracted from transfected
HEK293
cells and incubated on mica chips (Ted Pella, Inc) for 10 minutes. Samples
were then
rinsed twice with 100pIddH20 and left at RT to dry. The following day, atomic
force
microscopy was performed using a MFP-3D atomic force microscope (Asylum
Research).
lmmunofluorescence and Con focal microscopy
[0172] HEK293 cells transfected for co-culture experiments as
described
earlier were prepared for immunofluorescence and X-34 staining. After fixation
in 4%
paraformaldehyde for 15 min at RT, cells were washed twice in PBS at room
temperature (RT) for 5 min, and permeabilized in 0.25% Triton X-100 in PBS at
RT for
minutes. Cells were blocked with a blocking solution containing 1% normal goat

serum, 20 mg/ml BSA, 0.25% Triton X-100 in PBS for 3 h at RT. Primary mouse
monoclonal antibody against HA (Covance, Emeryville, CA) was diluted 1:2000 in

blocking solution and applied to cells overnight at 4 C. Cells were then
washed with
PBS containing 0.1% Triton X-100 3 times for 5 minutes each and incubated with
anti-
mouse A1exa546-conjugated secondary antibody (Invitrogen) diluted at 1:400 in
blocking solution. Cells were then washed with PBS containing 0.1% Triton X-
100 3
times for 5 min each, and exposed to 1 pM X-34 prepared in a solution of 40%
ethanol,
60% PBS, and 20 mM NaOH for 10 min at RT. Cells were then washed 3 times for 2

min each in 40% Et0H, 60% PBS and rinsed twice in 1X PBS for 5 min each.
Images
were captured using confocal microscopy (405 Confocal Microscope-Zeiss). For
the
characterization of the mechanism of HJ9.3 antibody blockade of propagation,
HEK293
cells were transfected with RD(AK)-YFP or mock transfected. Following culture
of
RD(AK)-YFP cells or mock-transfected cells in the presence of HJ9.3 for 48
hrs, cells
68

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
were fixed with 4% PFA, permeabilized with 0.25% TritonX-100 and then exposed
to
goat anti-mouse Alexa 546 labeled secondary antibody. Images were captured
using
confocal microscopy (Confocal Microscope- Zeiss).
Propidium Iodide (PI) cell death assay
[0173] HEK293 cells were plated at 75,000 cells/well in a 96-well
plate.
The following day, cells were transfected in quadruplicate with 100 ng of
various forms
of non-fluorescent RD-HA plasmids or exposed to transfection complexes without
DNA.
The next day, media containing transfection complexes were removed, and
replaced
with fresh media. Non-transfected cells were treated with varying
concentrations of
staurosporine (1, 2, 4, 20 pM) for 30 minutes at 37 C as a positive control
for cell death.
Staurosporine solution was then removed and all cells were exposed to 5 pg/ml
of
propidium iodide for 10 minutes at 37 C. Propidium iodide solution was then
replaced
with phenol-free media and fluorescence was read on the plate reader at 535nm
excitation and 617 nm emission.
lmmunoprecipitation
[0174] Transfected cell populations were co-cultured either alone
or in the
presence of mouse monoclonal antibody HJ9.3 (1:1000 which is equivalent to 2.5
ng/pl
of antibody) or pooled mouse IgG antibody for 3h, 6h, 9h, 12h, 24h or 48 h.
Conditioned
media were collected and protein-G-agarose beads (100 pl of 50% slurry beads
from
Pierce) were added to the media and incubated overnight at 4 C with rotation.
18 h
later, 500 pl of binding buffer (Pierce) was added to samples and centrifuged
at 2000 x
g for 3 minutes. Supernatant was discarded, and this wash step was repeated
three
times. Proteins bound to beads were then eluted using a high salt elution
buffer (50p1)
with incubation at room temperature for 5 minutes. Samples were then
centrifuged at
2000 x g for three minutes and supernatant collected. This elution step was
repeated
once for a total of 100pleluate. Another sample of conditioned media not
initially
exposed to HJ9.3 or IgG was incubated with the HJ9.3 (1:1000) or IgG
antibodies
overnight at 4 C with rotation, followed by the same immunoprecipitation
protocol as
69

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
described above. Samples from all conditions were analyzed on 4-20%
polyacrylamide
gels (BioRad) and detected with rabbit polyclonal antibody directed against
tau RD at
1:2000 dilution in 5% dry milk in TBS/Tween (ab64193, Abcam, Cambridge, MA). A

chemiluminescence-based peroxidaseconjugated secondary antibody reaction was
performed and detected by X-ray film.
Flow Cytometry
[0175] HEK293 cells were plated in a 10-cm plate at -80%
confluency.
Cells were then transfected with 24 pg of RD(LM)-YFP construct or transduced
with
mCherry lentivirus. The following day, cells were harvested by treating with
0.05%
trypsin for 3 minutes at 372C, pelleted and resuspended in fresh media. The
two cell
populations were co-cultured either alone or in the presence of mouse
monoclonal
antibody HJ9.3 directed against Tau-RD at 1:1000 or 1:10,000 dilutions for 48
h (1:1000
is equivalent to 2.5 ng/pl of antibody). After this time, cells were harvested
and
resuspended in Hanks balanced medium containing 1 /0 FBS and 1mM of EDTA.
Cells
premixed just prior to cytometry were used as negative controls. Cells were
counted
using the MoFlo high speed cell sorter (Beckman Coulter) and the percentage of
dual
positive cells was analyzed for each of the conditions. Each condition had
three
biological replicates, with 50,000 cells analyzed in each experimental
condition.
lntracerebroventricular (ICV) injection of anti-tau monoclonal antibodies
[0176] P3015 tau transgenic mice which express P3015 human T34
isoform (1N4R) were used in this study. At 6 months age these mice develop tau

pathology. Therefore, antibodies were infused into the left lateral ventricle
by
cerebroventricular injection at 6 months of age and these infusions were
carried for 12
weeks. After treatment, mice brains were processed for immunohistochemistry
and
biochemical analysis by ELISA and immunoblotting.
[0177] lntracerebroventricular injections were performed by using
Alzet
osmotic pumps, 2006 model. Brain cannula attached to an Alzet pump assembly
were
surgically implanted into the left lateral ventricle of each mouse at the
position 0.4 mm

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
anteroposterior to bregma, 1.0 mm lateral to midline and 2.5 mm dorsoventral.
After
treatment, placement of the cannula was verified by cresyl violet staining.
Introduction for Examples 9-15
[0178] Tau is a microtubule-associated protein that forms
intracellular
aggregates in several neurodegenerative diseases collectively termed
tauopathies.
These include Alzheimer's disease (AD), progressive supranculear palsy (PSP),
corticobasal degeneration (CBD), and frontotemporal dementia (FTD). Tau is a
highly
soluble and natively unfolded protein which binds and promotes the assembly of

microtubules. In tauopathies, tau accumulates in hyperphosphorylated
neurofibrillary
tangles (NFTs) that are visualized within dystrophic neurites and cell bodies
upon
appropriate staining. The amount of tau pathology correlates with progressive
neuronal
dysfunction and synaptic loss, and functional decline in humans and transgenic
mouse
models.
[0179] In human tauopathies, pathology progresses from one brain
region
to another in disease-specific patterns, although the underlying mechanism is
not yet
clear. The prion hypothesis holds that tau aggregates escape cells of origin
to enter
adjacent cells, where they seed further tau aggregation and propagate
pathology. The
inventors have previously observed that recombinant tau fibrils will induce
aggregation
of full-length intracellular tau in cultured cells, and that aggregated forms
of tau transfer
between cells (Frost et al., 2009; Nat Rev Neurosci 11, 155-159). Further, the
inventors
found that intracellular tau fibrils are released free into the media, where
they propagate
aggregation by direct interaction with native tau in recipient cells. An anti-
tau antibody
(HJ9.3) blocks this process by preventing tau aggregate uptake into recipient
cells
(Kfoury et al., 2012; J Biol Chem 287, 19440-19451). In addition to similar
experiments
with recombinant tau, it has been shown that paired helical filaments from AD
brain
induce cytoplasmic tau aggregation. Injection of brain extract from human
P301S tau
transgenic mice into the brains of mice expressing wild-type human tau induces

assembly of wild-type human tau into filaments and spreading of pathology.
Similar
effects occurred after injection of recombinant full-length or truncated tau
fibrils, which
71

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
caused rapid induction of NFT-like inclusions that propagated from injected
sites to
connected brain regions in a time-dependent manner. Finally, selective tau
expression
in the entorhinal cortex caused late pathology in the axonal terminal zones in
cells in the
dentate gyrus and hippocampus, consistent with trans-synaptic movement of
aggregates. A growing body of work thus supports the idea that tau aggregates
transfer
between cells, and might be targeted with therapeutic antibodies.
[0180] In mouse models that mimic aspects of AD and Parkinson's
disease (PD), passive immunization using antibodies against A13 and alpha
synuclein
can reduce Ar3 and alpha-synuclein deposition in brain, and improve behavioral
deficits.
Active immunization in tauopathy mouse models using tau phospho peptides
reduced
tau pathology and in some studies improved behavior deficits. However, in one
study
active immunization of C57BL/6 wild type mice with full length recombinant tau
induced
tau pathology and neurologic deficits. In two passive vaccination studies,
there was
reduced tau pathology and improved motor function when the antibody was given
prior
to the onset of pathology. While several of the tau immunization studies
appear to have
some beneficial effects, the maximal expected efficacy of anti-tau antibodies
administered after the onset of pathology, the optimal tau species to target,
and the
mechanism of the therapeutic effect have remained unknown.
Example 9. Characterization of anti-tau antibodies
[0181] The inventors have previously observed that tau aggregates,
but
not monomer, are up taken by cultured cells, and that internalized tau
aggregates
trigger intracellular tau aggregation in recipient cells (Frost et al., 2009;
Nat Rev
Neurosci 11, 155-159; Kfoury et al., 2012; J Biol Chem 287, 19440-19451). The
HJ8
series of 8 mouse monoclonal antibodies (raised against full-length human tau)
and HJ9
series of 5 antibodies (raised against full-length mouse tau) were
characterized in an
adapted cellular biosensor system previously described in Kfoury et al. (2012;
J Biol
Chem 287, 19440-19451) that measures cellular tau aggregation induced by the
addition of brain lysates containing tau aggregates. The antibodies had
variable effects
in blocking seeding, despite the fact that all antibodies efficiently bind tau
monomer and
72

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
stain neurofibrillary tangles. Three antibodies were selected with different
potencies in
blocking seeding for the studies presented herein.
[0182] Prior to testing in vivo, the binding affinities and
epitopes of the
antibodies, which are all IgG2b isotype, were determined. Human and mouse tau
was
immobilized on a sensor chip CM5 for surface plasmon resonance (SPR) (FIG.
30). The
HJ9.3 antibody, raised against mouse tau, recognizes both human (FIG. 30A) and

mouse (FIG. 30B) tau with the same binding constant (KD = Kd/K, = 100 pM)
(FIG.
30G). The association (Ka) and dissociation (Ka) was calculated by using
BlAevaluation software (Biacore AB) selecting Fit kinetics simultaneous Ka/Kd
(Global
fitting) with 1:1 (Langmuir) interaction model. The Ka and Kd of HJ9.3 towards
human
(K, = 7.5 x 104 Ms-1, Kd = 7.5 x 10-6 s-1) and mouse tau (Ka = 8.6 x 104 Ms-1,
Kd = 9.1 x
10-6 s-1) indicate strong binding to both. The epitope of HJ9.3 was mapped to
the
repeat domain (RD) region, between amino acids 306-320. HJ9.4, raised against
mouse
tau, had high affinity KD (2.2 pM) towards mouse tau with a high association
rate
constant (K, = 2.28 x 105 Ms-1) and very low dissociation constant (Kd = 5.1 x
10-7 s-1)
(FIG. 300 and Table 4. However, the same antibody had a much lower affinity
(KD = 6.9
nM) toward human tau (FIG. 30C and Table 4) with a similar association rate
constant
(K, = 1.5 x 105 Ms-1) as with mouse tau but with much faster dissociation (Kd
= 1.07 x
10-3 s-1). Thus, the HJ9.4 interaction with human tau is less stable than with
mouse tau.
The epitope for this antibody is amino acids 7-13. HJ8.5 was raised against
human tau.
It binds to human tau (FIG. 30E) but not to mouse tau (FIG. 30F). The KD (0.3
pM) (FIG.
30E and Table 4) and low dissociation rate (Kd = 4.38 x 10-8 S-1), indicate
that HJ8.5
binds human tau with very high affinity. The epitope of HJ8.5 was mapped to
amino
acids 25-30. All 3 anti-tau antibodies strongly recognized human tau fibrils
on SPR (FIG.
31). Because the fibrils have multiple identical epitopes, the association and
dissociation rates could not be directly calculate.
73

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Table 4 Association rate constant (Ka), dissociation rate constant (Ka) and
binding
constant (KD) of each antibody towards human and mouse tau. BlAevaluation
software
(Biacore AB) was used to calculate Ka and Kd by selecting Fit kinetics
simultaneous
Ka/Ka (Global fitting) with 1:1 (Langmuir) interaction model. Ms-
1=millisecond, M=molar,
s=second
11,19.3 I-1194 11J8.5
(Me) 755x 104 1 53 x 105 3 x
(s-) 7,51 x 11I6 1.07 x 10 4.34x 1Os
Ko (M)
99 pM 6.9 nM 0.336 pM
g Ka (Ms) 8.61 x 104 2.28 x 105
0 Kd (s-) 916 x 1O 5.1 x 10 '
0
2
K( M} 100 pM 2.24 pM
[0183] The antibodies were also assessed by immunoblotting and
immunostaining. On Western blots, all 3 antibodies bound to human tau (FIG.
30H).
HJ9.3 and HJ9.4 bound to mouse tau while HJ8.5 did not (FIG. 30H). Consistent
with
our prior findings of the inventors (Yamada et al., 2011; J Neurosci 31, 13110-
13117),
there appeared to be less reassembly buffer (RAB) soluble tau in 9 month old
compared
to 3 month old P301S mice. It was also found that HJ8.5 stained human tau in 3
month
and 9-12 month old transgenic P301S mouse brains. Tau immunoreactivity was
present
throughout the cell bodies and processes (FIG. 32). In 9-12 month old P301S
mice with
tau aggregates, HJ8.5 detected tau aggregates in cell bodies (FIG. 32A). Other

antibodies produced similar results (Table 5). All antibodies bound to
neurofibrillary
tangles and neuropil threads in AD brain (FIG. 32).
74

CA 02877397 2014-12-18
WO 2014/008404
PCT/US2013/049333
Table 5. Relative efficacy of anti-tau antibodies in different assays.
HJ8.5 HJ9.3 HJ9.4
Method Human Mouse Human Mouse Human Mouse
tau tau tau tau tau tau
Western blot ++ - +++ +++ ++ ++
lmmunostaining +++ + + ++ +++
Human AD +++ N/A + N/A + N/A
brain NFT's
Example 10. Tau-antibodies block the uptake and seeding activity of P301S tau
aggregates
[0184] To evaluate seeding activity present in P301S brain lysates,
a
cellular biosensor system previously described by the inventors (Kfoury et
al., 2012)
was adapted. This is based on expression of the repeat domain of tau (aa 243-
375)
containing the AK280 mutation fused either to cyan or yellow fluorescent
protein
(RD(AK)-CFP /YFP). Uptake of exogenous aggregates into these cells triggers
intracellular aggregation of RD(AK)-CFP/YFP that is detected by fluorescence
resonance energy transfer (FRET) recorded on a fluorescence plate reader.
Clarified
brain lysates from 12 month old P301S mice added to the biosensor cell system
induced strong aggregation of the RD(AK)-CFP/YFP reporter, indicating the
presence of
tau seeding activity (FIG. 33A). The seeding activity from 12-mo P301S brain
homogenate mice roughly corresponds to 50 nM (monomer equivalent) of
recombinant
full length fibrils (data not shown).
[0185] There was little to no aggregation induced by lysates from
tau
knockout mice, wild-type mice, or 3 month old P301S mice lacking tau pathology
(FIG.
33A). The anti-tau antibodies (HJ8.5, HJ9.3 and HJ9.4) were assessed for their
ability
to block the uptake, and seeding activity of these lysates. HJ3.4 (mouse
monoclonal
anti-An antibody) was a negative control. The anti-tau antibodies effectively
blocked
seeding activity (FIG. 33B). To determine their relative efficacy, the
antibodies (0.125,
0.25, 0.5, 1, 2 pg/ml) were titrated against a fixed amount of P3015 brain
lysate (FIG.
33C). The HJ8.5 antibody blocked seeding activity at concentrations as low as
0.25
pg/m1 compared to controls. At 0.5 pg/ml, both HJ8.5 and HJ9.3 antibody
significantly
blocked uptake and seeding activity compared to control. HJ9.4 was least
potent in

CA 02877397 2014-12-18
WO 2014/008404
PCT/US2013/049333
blocking the uptake and seeding activity, consistent with its higher affinity
for mouse tau.
All 3 anti-tau antibodies detected tau aggregates internalized following
uptake by
HEK293 cells, as detected by post-hoc cellular permeabilization and staining.
However,
when these antibodies were pre-incubated with and without P301S brain lysates,
none
of these antibodies were detected inside cells upon staining with anti-mouse
secondary
antibody (FIG. 34). While other modes of inhibition are possible, these data
are
consistent with a mechanism based on blocking cellular uptake of tau
aggregates.
Example 11. Intracerebroventricular infusion of anti-tau antibodies
[0186] In
the mouse colonies, P3015 mice first develop intracellular tau
pathology beginning at 5 months of age. To test the efficacy of the 3
antibodies by
chronic intracerebroventricular (ICV) administration, a catheter was
surgically implanted
into the left lateral ventricle of each mouse at 6 months of age and
continuously infused
anti-tau antibodies for 3 months via Alzet subcutaneous osmotic mini-pump
(FIG. 35A).
Anti-A8 antibody HJ3.4 and phosphate buffered saline (PBS) were used as
negative
controls. After 6 weeks, each pump was replaced with one filled with fresh
antibody
solution or PBS. At the time of brain dissection, catheter placement in the
left lateral
ventricle of each mouse was verified by cresyl violet staining (FIG. 35B).
Only mice with
correctly placed catheters were included in the analyses. To test the
stability of the
antibodies after 6 weeks in vivo (FIG. 35A), residual pump contents were
collected upon
removal from the animals, and the antibodies were assessed using SDS-PAGE and
Coomassie blue staining. Light and heavy chains were intact, with no
fragmentation,
and retained tau binding activity on western blot (data not shown). To
estimate the
concentration of anti-tau antibodies in CSF and serum during the infusion,
biotinylated
HJ8.5 (HJ8.5B) was administered for 48 hours (-7.2 rig/day) (FIG. 35A). The
concentration of free HJ8.5B was 7.3 pg/m1 in the CSF and 6.2 pg/m1 in the
serum,
indicating significant clearance of the antibody from the CNS to the periphery
(Table 6).
HJ8.5B bound to human tau was also detected in both CSF and serum, though the
concentration was lower than that of free antibody (Table 6).
76

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Table 6. Levels of biotinylated HJ8.5 antibody that is free (not bound to tau)
and HJ8.5
antibody bound to tau in serum and cerebrospinal fluid (CSF) 48 hrs after IF
or ICV
administration.
Conc. Of free HJ8.5B (.1g/m1)
Treatment CSF Serum
CSF Comc. As
% Serum Conc.
HJ8.5B injected IF 0.9 0.1 552 38.6 0.16 0.02
(50mg/kg/48hrs)
HJ8.5B injected ICV 7.3 1.6 6.2 0.5 95.4 19.4
(ca. 14 g/48hrs)
Conc. Of HJ8.5B bound to tau (.1g/m1)
HJ8.5B injected ICV 0.10 0.02 0.04
53
(ca. 14 g/48hrs) +
4.6
0.03
Example 12. Anti-tau antibody treatment reduces abnormally phosphorylated tau
[0187] To determine the extent of tau pathology in P301S mice after
3
months of treatment, multiple stains for tau pathology were carried out. Brain
sections
were first assessed by immunostaining with the anti-phospho tau antibody AT8
(FIG.
36). AT8 binds phosphorylated residues Ser202 and Thr205 of both mouse and
human
tau (FIG. 36). In mice treated with PBS and HJ3.4, AT8 strongly stained
neuronal cell
bodies and the neuropil in multiple brain regions, particularly in the
piriform cortex,
entorhinal cortex, amygdala, and hippocampus (FIG. 36A and 36B). HJ8.5
treatment
strongly reduced AT8 staining (FIG. 36C), especially in the neuropil. HJ9.3
and HJ9.4
also decreased AT8 staining but the effects were slightly less (FIG. 360 and
36E).
Quantitative analysis of AT8 staining in piriform cortex (FIG. 37A),
entorhinal cortex
(FIG. 37B), and amygdala (FIG. 37C) demonstrated a strong but variable
reduction in
phospho-tau in all anti-tau antibody treated mice. HJ8.5 antibody markedly
reduced AT8
staining in piriform cortex, entorhinal cortex, and amygdala. HJ9.3 had
slightly
decreased effects compared to HJ8.5, and HJ9.4 had significant effects in both

entorhinal cortex and amygdala but not in the piriform cortex (FIG. 37). The
hippocampus exhibited much more variable AT8 staining vs. other brain regions,

predominantly in cell bodies, and thus was not statistically different in
treatment vs.
control groups (FIG. 370). Because it has been reported that male P301S mice
have
greater tau pathology than females, the effect of both gender and treatment
were also
77

CA 02877397 2014-12-18
WO 2014/008404
PCT/US2013/049333
assessed (FIG. 38). In addition to an effect of treatment, there was
significantly more
AT8 staining in all brain regions analyzed in male mice (Table 7). However,
the effects
of the antibodies were still highly significant and virtually identical after
adjusting for
gender (Table 8). The treatment groups versus controls in males and females
were also
compared separately, and the effects of antibody HJ8.5 remained most
significant (FIG.
38A and 38B).
Table 7. p Values of Treatment/Gender
Amygdala Entorhinal Hippocampus
Piriform
cortex cortex
Treatment 0.0107 0.0053 0.2917 0.0147
Gender 0.0026 0.0027 0.0244 0.0067
p values determined by two-way ANOVA considering treatment and gender as
factors.
For amygdala, entorhinal cortex, and piriform cortex regions, treatment and
gender are
both significant factors with p values <0.05, but for hippocampus CA1 region,
treatment
is not a significant factor with p value =0.2917 while gender is a significant
factor with p
value=0.0244.
Table 8
Amygdala Entorhinal Hippocampus Piriform cortex
cortex CA1
P P P P p p p p
value-1 value-2 value-1 value-2 value-1 value-2 value-1 value-2
Control 0.0009 0.0009 0.0022 0.0022 0.0421 0.0526 0.011 0.0113
vs.
HJ8.5
Control 0.0956 0.1605 0.0335 0.0576 0.2486 0.3889 0.0566 0.0982
vs.
HJ9.3
Control 0.0106 0.0072 0.0077 0.005 0.2427 0.2427 0.1787 0.1569
vs.
HJ9.4
p values were calculated before and after adjustment of gender. p value-1: not
adjusted
by gender; p value-2: adjusted by gender. p value-1 was determined by one-way
ANOVA, treatment is the independent variable. p value-2 was determined by two-
way
ANOVA, treatment and gender are independent variables.
78

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Example 13. Correlation of multiple staining modalities
[0188] To test for tau amyloid deposition, thioflavin S (ThioS) was
used to
stain brain sections (FIG. 39). ThioS staining was semi-quantitatively
assessed using a
blinded rater who gave a score from 1 (no staining) to 5 (maximum staining) in
all
control and anti-tau antibody treated mice. By semi-quantitative assessment,
HJ8.5
treatment significantly reduced ThioS staining compared to PBS and HJ3.4 (FIG.

39A and 39B). Mice treated with PBS, HJ8.5, and HJ9.3 (n=6 from each group)
were
also stained with PHF1 monoclonal antibody, which recognizes tau phospho-
residues
5er396 and 5er404. AT8 and PHF1 staining significantly correlated (r = 0.630,
p =
0.005) (FIG. 40A) showing that 2 anti-phospho tau antibodies to different tau
epitopes
give similar results.
[0189] Many neurodegenerative diseases, including tauopathies,
exhibit
microglial activation in areas of the brain surrounding protein aggregation
and cell injury.
Microglial activation was assessed in the treatment groups using anti-CD68
antibody
(FIG. 41). HJ8.5 and HJ9.3 treatment reduced microglial activation in piriform
cortex,
entorhinal cortex, and amygdala compared to controls (FIG. 41A-41D). HJ9.4 had
a
weaker effect in the piriform cortex compared to HJ8.5 and HJ9.3 (FIG. 41C-
41E),
consistent with the AT8 staining results (FIG. 37A). Microglial activation
strongly
correlated with AT8 staining in all samples (r = 0.511, p = 0.0038) (FIG.
40B).
Example 14. Anti-tau antibodies reduce detergent-insoluble tau and seeding
activity
[0190] To determine the level of soluble and insoluble tau in the
cortex,
sequential biochemical extraction with RAB (aqueous buffer), radio
immunoprecipitation
assay (RIPA)(detergent buffer), and 70% formic acid (FA) were performed to
solubilize
the final pellet. Total tau was quantified by ELISA with anti-tau antibody
HJ8.7, which
detects both human and mouse tau with the same KD (0.34 pM). The possibility
that the
treatment antibodies would interfere with the ELISA was excluded by spiking
positive
control samples with these antibodies prior to analysis and observing no
interference
(data not shown). All mice that were assessed by pathological analysis in FIG.
37 were
analyzed. Total tau levels in the RAB (FIG. 42A) or RIPA (FIG. 42B) soluble
fractions
79

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
were similar among all groups. The detergent-insoluble/70% FA soluble
fractions were
analyzed by neutralizing the samples prior to ELISA and western blot. Every
animal
studied was analyzed, and it was found that HJ8.5 and HJ9.3 decreased
detergent-
insoluble tau by >50% vs. controls (FIG. 42C). Representative samples (n=4
from each
group) illustrate by western blot decreased levels of insoluble tau in mice
treated with
HJ8.5 and HJ9.3 (FIG. 40C). Insoluble tau levels were no different in HJ9.4-
treated
groups versus PBS or HJ3.4. Human and mouse tau were also assessed
specifically in
the detergent-insoluble/70% FA soluble fractions in N=6 mice per group in
which the
mean AT8 staining reflected the mean values of results in FIG. 37. There was
significantly more human tau than mouse tau in the 70% FA soluble fraction,
and the
antibodies significantly lowered human but not mouse tau in this fraction
(FIG. 420 and
42E). In these same samples, levels of AT8 immunoreactive signal were assessed
by
ELISA. The AT8 signal was lower in the antibody treated samples (FIG. 42F),
similar to
what was seen for total tau in this fraction.
[0191] It was hypothesized that a reduction of tau aggregation in
brain
would correlate with a reduction in seeding activity. Thus, the cellular
biosensor assay
was used to test for P3015 brain seeding activity in the cortical RAB soluble
fractions
from the different treatment groups. Prior data by the inventors assessing ISF
tau in
P3015 mice suggested the possible presence of extracellular tau aggregates in
equilibrium with both the biochemically soluble and insoluble pools of tau
(Yamada et
al., 2011; J Neurosci 31, 13110-13117). First, intracellular aggregation of
RD(AK)-
CFP/YFP was assessed after treating the cells with lysates from mice treated
with PBS
or HJ3.4. Lysates from these groups strongly induced FRET signal (FIG. 43A).
Markedly less seeding activity was observed in lysates from the cortical
tissue of mice
treated with HJ8.5 and HJ9.3 (FIG. 43A). This was not due to residual antibody
in the
brain lysates, because immunoprecipitation of the brain lysates followed by
elution of
seeding activity from the antibody/bead complexes produced the same pattern
(FIG.
43B). Thus HJ8.5 and HJ9.3 reduce seeding activity in the P3015 tau transgenic
mouse
brain. HJ9.4 did not significantly reduce seeding activity (FIG. 43A). Seeding
activity
strongly correlated with the amount of detergent-insoluble/formic acid-soluble
tau

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
detected by ELISA (Pearson's r = 0.529, p = 0.0001) (FIG. 43C), but did not
correlate
with total tau in RAB fractions (FIG. 430). It was hypothesized that seeding
activity is
due to tau aggregates present in the RAB soluble fraction. To test for this,
Semi-
Denaturing Detergent-Agarose Gel Electrophoresis (SDD-AGE) was performed
followed
by Western blot. In addition to tau monomer, higher molecular weight tau
species
present in 3 month old P3015 mice and a larger amount present in 9 month old
P3015
mice was observed (FIG. 43E). A component of these higher molecular weight
species
likely constitutes the seeding activity detected in the FRET assay and may be
in
equilibrium with the tau present in the detergent-insoluble/formic acid-
soluble fraction.
Example 15. Anti-tau antibodies rescue contextual fear deficits
[0192] In studies of P301S Tau transgenic mice at 9 months of age,
the
control and anti-tau antibody treated groups were compared in a variety of
behaviors.
The groups did not differ in locomotor activity, exploration, or measures of
sensorimotor
function (FIG. 44). The ability of the anti-tau antibody treatments to rescue
cognitive
deficits in P3015 mice was evaluated by assessing the performance of the mice
on the
conditioned fear procedure. On day 1, all four treatment groups of mice
exhibited similar
levels of baseline freezing during the first two minutes in the training
chamber. This was
confirmed by rmANOVA, which failed to reveal any significant overall main
effects or
interactions involving treatment (FIG. 45A). In addition, all four groups
showed similar
levels of freezing during the tone-shock (T/S) conditioned stimulus-
unconditioned
stimulus (CS-US) pairings (FIG. 45A). The general lack of differences in
freezing levels
between groups across the three T/S pairings was documented by a non-
significant
effect of Treatment and a non-significant Genotype by Minutes interaction.
[0193] In contrast to the absence of differences among groups
during
testing on day 1, there were robust differences in freezing levels from the
contextual
fear test (form of associative learning) conducted on day 2 between two of the
anti-tau
antibody groups and the PBS+HJ3.4 control mice (FIG. 45B). Subsequent planned
comparisons indicated that the HJ8.5 mice showed significantly elevated
freezing levels
averaged across the 8-minute test session (FIG. 45C) compared to the PBS+HJ3.4
81

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
control group, [F(1,45)=8.30, p=0.006], as did to a lesser extent the HJ9.4
mice,
[F(1,45)=5.60, p=0.022]. Thus, HJ8.5 appeared to have a stronger effect
overall in
preserving associative learning.
Discussion for Examples 9-15
[0194] One model for the pathogenesis of the tauopathies holds that
aggregates produced in one cell escape or are released into the extracellular
space to
promote aggregation in neighboring or connected cells. It was observed that
selection
of therapeutic antibodies that specifically block tau seeding activity from
brain lysates
predicts potent in vivo responses at least as strong if not stronger than
prior reports of
active or passive tau vaccination. Experiments were began with a cellular
biosensor
assay that is sensitive to the presence of extracellular tau aggregates. It
was found that
brain lysates from P301S transgenic mice contained seeding activity that could
induce
further intracellular aggregation. After screening a panel of anti-tau
antibodies, three
were selected with variable activities in blocking tau seeding activity. These
antibodies
were infused ICV over three months into P301S tauopathy mice, beginning at a
time
when pathology had initiated (6 months). Infusion of the antibodies resulted
in
appreciable concentrations of antibody present in both CSF and serum,
consistent with
previous reports of efflux of antibodies from the CNS to the periphery.
Treatment with
HJ8.5, the most potent antibody in vitro, profoundly reduced tau pathology,
strongly
decreasing it from the neuropil. This effect was detected with multiple
independent
stains, biochemical analyses of insoluble tau, and by analysis of residual tau
seeding
activity present in brain lysates. Further, this treatment improved the one
behavioral
deficit detected in this model. All antibodies block tau aggregate uptake into
cells, and
none is observed within cells in the presence or absence of extracellular
aggregates in
the assays. The efficacy of these antibodies implies a clear role for
extracellular tau in
the pathogenesis of neuropathology that was previously thought to be cell-
autonomous.
Further, this work extends prior findings by the inventors, which suggest that
aggregate
flux may occur in the setting of intracellular pathology, raising the
possibility of therapies
that can assist in aggregate clearance by targeting extracellular species.
Finally, this
82

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
work has important implications for the design of therapeutic antibodies, and
suggests
that targeting seeding activity in particular may produce the most effective
agents.
[0195] Mechanism-based antibody therapy Several prior active and
passive peripheral immunotherapy approaches against tau have also reduced tau
pathology and improved behavioral deficits, but the underlying rationale for
antibody
choice was based either on a phospho-epitope, reactivity with neurofibrillary
tangles, or
was not stated. One tau immunization study, performed by vaccinating mice with
full
length tau, induced pathology in wild type mice. However, subsequent active
immunization approaches with phospho-tau peptides in tau transgenic models
reduced
tau pathology and showed behavioral improvement. In a passive immunization
study,
JNPL3 tau transgenic mice were administered the PHF1 antibody
intraperitoneally at 2-
3 months of age, prior to the onset of tauopathy. PH F-1 targets a
pathological form of
abnormally phosphorylated tau. Treatment reduced tau pathology and improved
behavior. However, while it decreased insoluble phosphorylated tau, total
insoluble tau
did not change. In another passive immunization study, JNPL3 and P3015 mice
(at age
2-3 months, prior to the onset of tauopathy) were peripherally administered
the PHF1 or
MC1 antibody, which targets an aggregate-associated epitope. Both treatments
improved tau pathology and delayed the onset of motor dysfunction. In these
prior
studies, the mechanism of action of the antibodies was not clear, and none was

explicitly tested. Indeed, some proposed an intracellular mechanism. Moreover,
no
study appears to have produced the magnitude of reduction in tau pathology
described
in the examples provided herein, with the caveats that antibodies were infused
into the
CNS while the other studies utilized peripheral infusion; and different animal
models
were utilized.
[0196] This study was explicitly designed to test a prediction that
extracellular tau seeds are a key component of pathogenesis. The study began
with a
selection process to pick antibodies capable of blocking tau seeding in vitro,
purposely
testing agents with a range of predicted activities. All antibodies tested in
vivo effectively
block aggregate uptake and seeding, providing a basis for their observed
activity. In
addition, correlation of antibody affinity, epitope, isotype, glycosylation,
and ability to
83

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
bind phosphorylated forms of tau may be important to assess in future studies.
This is
also the first study to report the effects of direct, intra-CNS infusion of
anti-tau
antibodies. Despite the fact that the antibodies utilized each target
different tau epitopes
and none targets phospho-tau, 2 of 3 strongly reduced abnormal tau load both
immunohistologically and biochemically, and two significantly improved memory,
one to
a greater extent than the other. Effects on tau pathology also correlated very
well with a
reduction in intrinsic seeding activity.
[0197] HJ8.5 and HJ9.3 strongly decreased pathological tau seeds in
vivo.
A strong reduction in tau pathology might occur by preventing induction of tau

aggregation in neighboring cells. While HJ9.4 did not decrease pathology as
potently, it
did decrease tau pathology in the amygdala. The variation in effectiveness in
different
brain regions among the antibodies may be due to the formation of region-
specific
aggregate conformers for which the antibodies have subtle differences in
binding
affinity.
[0198] Once extracellular tau aggregates are sequestered by anti-
tau
antibodies in vivo, their metabolic fate is not yet clear. After 3 months of
antibody
administration, reduced microglial activation were found, presumably due to
less tau-
related pathology and neurodegeneration. However, this could be due to more
efficient
clearance of extracellular aggregates, with a reduction in related microglial
activation.
Several months of passive immunization with anti-A8 antibodies has also been
noted to
reduce microgliosis. The mechanism by which antibody/tau complexes are cleared
in
vivo, and the mechanism via which they decrease tau pathology, remains to be
definitively clarified. It has been suggested that immunization with anti-a-
synuclein
antibodies clears a-synuclein aggregates by promoting lysosomal degradation. A
recent
study with anti-a-synuclein antibodies showed that the antibodies targeted a-
synuclein
clearance mainly via microglia, presumably through Fc receptors. Neurons
express Fcy
receptors, and may be able to internalize IgG complexed with antigen by high
affinity
FcyRI receptor. Internalized tau antibodies may also contact tau in endosomes
and
eventually induce clearance of intracellular tau aggregates by the
endosomal/autophagy-lysosomal system. Though the anti-tau antibodies used in
the
84

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
study described herein can bind extracellular tau assemblies, no evidence of
significant
localization within cells was found. That does not, however, rule out the
possibility that
cells in vivo take up antibody/tau complexes to influence tau aggregate
clearance as
well as inflammation. For example, it has recently been shown that antibodies
complexed with viruses can bind to the cytosolic IgG receptor TRIM21,
targeting the
antibody/virus complex to the proteasome. In addition, antibodies bound to
TRIM21
were shown to activate immune signaling. While interaction with antibodies/non-

infectious antigen complexes with TRIM21 has not yet been shown, it may be
interesting to determine if such a mechanism is relevant to the anti-tau
antibodies.
Interestingly, there is also evidence in the P301S model of tauopathy that the
innate
immune system is activated prior to the development of significant tau
pathology, and
that early immunsuppresion attenuates tau pathology. It may be possible that
antibodies
capture tau aggregates induced by inflammation, reducing subsequent aggregate-
induced inflammation and disease progression.
[0199] Extracellular tau and spreading of tau pathology The work
presented herein implicitly tests the role of extracellular tau in
pathogenesis. It is now
abundantly clear that extracellular tau aggregates can trigger fibril
formation of native
tau inside cells, whether their source is recombinant protein or tau extracted
from
mammalian cells. A role for free tau aggregates was originally hypothesized
(i.e. not
membrane-enclosed) as mediators of trans-cellular propagation based on our
prior
work, in which HJ9.3 added to the cell media blocked internalization, and
immunoprecipitated free fibrils (Kfoury et al., 2012; J Biol Chem 287, 19440-
19451).
[0200] In animal models, tau aggregates can apparently spread from
one
region to another (e.g. entorhinal cortex to neurons downstream in the dentate
gyrus
and hippocampus). The inventors have found that monomeric tau is constantly
released
in vivo into the brain interstitial fluid even under non-pathological
conditions (Yamada et
al., 2011; J Neurosci 31, 13110-13117). The inventors also found that
exogenous
aggregates would reduce levels of soluble ISF tau, suggesting that seeding
and/or
sequestration phenomena can occur in this space (Yamada et al., 2011; J
Neurosci 31,
13110-13117). Taken together, abundant evidence supports the concept that

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
extracellular tau aggregates form, and can be taken up by adjacent cells,
connected
cells, or possibly back into the same cell, thereby increasing the burden of
protein
misfolding. This evidence makes a clear prediction: therapy that captures
extracellular
seeding activity should ameliorate disease. The findings described in the
examples
presented herein are consistent with this idea.
[0201] The role of tau flux in pathogenesis It would not be
predicted a
priori that a mouse model such as P301 S, which drives mutant tau expression
via the
prion promoter in virtually all neurons, should benefit from antibody
treatments that
block trans-cellular propagation of aggregation. In theory, pathology could
occur
independently in all neurons that express this aggregation-prone protein.
However,
prior work by the inventors in tissue culture suggested a role for flux of tau
aggregates,
since HJ9.3 added to the cell media increased the steady state level of
aggregates over
time. While the model of aggregate flux requires further testing, the results
presented
herein are consistent with this idea, since antibody treatment profoundly
reduced
intracellular tau pathology. It is predicted that antibodies that block tau
uptake may
create a "sink" in the extracellular space that may promote clearance by
another
mechanism, possibly involving microglia.
[0202] Therapeutic antibodies and targeting seeding activity The
pharmaceutical industry is devoting increasing efforts to develop therapeutic
antibodies
that target aggregation-prone proteins that accumulate within cells. The
principal criteria
have been that the antibodies will bind epitopes known to accumulate in
diseased brain.
This approach may or may not lead to antibodies with optimal activity in vivo.
The
examples herein supports a new model of therapeutic antibody development that
emphasizes efficacy in blocking the seeding activity present in the brain.
Using this
approach, antibodies with higher apparent efficacy than has previously been
reported
were identified. In an extension of the prion hypothesis, it is further
proposed that
distinct tau aggregate "strains" may predominate in patients with different
types of
tauopathy, and these may have unique sensitivities to different antibodies. In
any case,
the use of sensitive in vitro assays of antibody efficacy as described here
may allow
much more efficient development and optimization of antibody-based therapies.
86

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
[0203] The strong protective effect of the anti-tau antibodies,
particularly
with the HJ8.5 antibody, suggests that this type of approach should be
considered as a
treatment strategy for human tauopathies. While chronic administration of
antibodies via
an ICV approach may be possible, in future studies, it may be important to
determine
the PK/PD response with peripheral administration of these antibodies when
given in
both a prevention and treatment mode. In addition, the tau seeding assay may
be useful
to monitor target engagement by the antibodies.
Experimental Procedures for Examples 9-15
[0204] Antibodies HJ9.3 and HJ9.4 mouse monoclonal antibodies were
raised by immunizing tau knockout mice (The Jackson laboratory) against mouse
tau,
and HJ8.5 and HJ8.7 monoclonal antibodies were raised by immunizing tau
knockout
mice against human tau. HJ9.3, HJ9.4 and HJ8.7 monoclonal antibodies recognize
both
mouse and human tau. However, HJ8.5 monoclonal antibody binds only to human
tau
(epitope is at residues 25-30 [NCBI reference sequence: NP_005901]). HJ9.3
antibody
recognizes the RD region of tau (epitope at residues 306-320). HJ9.4 antibody
recognizes the N-terminal region of tau (epitope is at residues 7-13). As a
control
antibody, HJ3.4 mouse monoclonal antibody was used, which recognizes the N-
terminal
region of the human A6 sequence (epitope at residues 1-16). HJ8.5, 9.3, and
9.4
monoclonal antibodies are of the IgG2b isotype. Rabbit polyclonal tau antibody

(ab64193, epitope located at repeat domain region) was purchased from Abcam.
Mouse
monoclonal biotinylated BT-2 antibody, recognizes human and mouse tau (epitope
at
residues 194-198) and was purchased from Pierce. Rat anti-mouse monoclonal
CD68
antibody was purchased from AbD SeroTec. Biotinylated AT8 antibody was
purchased
from Thermo scientific.
[0205] Surface plasmon resonance Surface plasmon resonance
experiments were performed on BlAcore 2000 surface plasmon resonance
instrument
(GE Healthcare-BlAcore). Biacore sensor chip CM-5 was activated by using EDC
(1-
ethy1-3-(3-dimethylaminopropy1)-carbodiimide) and NHS (N-hydroxysuccinimide)
in a
1:1 ratio for 7 minutes. The sensor chip surface was saturated by immobilizing
5 pg/m1
87

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
of recombinant human or mouse tau or human tau fibrils in 10 mM Sodium acetate
pH
3.5 with a flow rate of 5 I/min. The remaining unbound area was blocked by 1
M
Ethanolamine pH 8.5. For reference, one flow cell is activated with NHS and
EDS,
followed by blocking with 1M ethanolamine. Then all the anti-tau antibodies
were
injected at different concentrations (0.11, 0.23, 0.46, 0.9, 1.8, 3.7, 7.5
g/ml) in filtered,
degassed 0.01 M Hepes buffer, 0.15 M NaCI, 0.005% surfactant P20, pH 7.4 at a
flow
rate of 10 I/min. All samples were run in duplicate. After each run with a
single
antibody concentration, the surface of the chip was totally regenerated by
using 10 mM
Glycine pH 1.7, to remove the bound antibody to tau, without disturbing the
immobilized
tau on the chip. Data analysis was performs by using BlAevaluation software
(GE
healthcare-BlAcore).
[0206] Tau fibrilization 8 M recombinant full length human tau was
pre-
incubated with 2mM dithiothreitol for 45 min at room temperature then 10 mM
HEPES
and 100mM NaCI and 8 M heparin were added for a total volume of 200 I
followed by
incubation for 7 day at 37 C to form fibrils. After fibril formation, the
remaining
monomers of tau in the sample were separated by using 100 kDa Microcon
centrifugal
filters according to manufacturer's instructions (Millipore).
[0207] IP and ICV administration of biotinylated HJ8.5 antibody
Mouse
monoclonal HJ8.5 antibody was biotinylated according to the manufacturer's
instructions (Sulfo-NHS-LC-Biotin kit, Pierce). Biotinylated HJ8.5 (HJ8.5B)
was
administered by interperitoneal injection (IP) at 50mg/kg in 5-6 month old
P301S mice
(n=3). After 48 hrs, mice were sacrificed. Serum and CSF was collected and
stored at -
80 2C until use. HJ8.5B was also administered by intracerebroventricular
injection (ICV)
by surgically implanted osmotic pumps into the left lateral ventricle of 5-6
month old
P3015 mice (n=3). This antibody was continuously infused for 48 hrs. After 48
hrs, mice
were sacrificed. Serum and CSF was collected and stored at -80 2C until use.
[0208] lnracerebroventricular (ICV) injection procedure ICV
infusions were
performed by Alzet osmotic pumps, 2006 model (Durect). The age of the mice was
6
months at the time of surgery. Before the surgery, an L-shaped cannula was
attached
to tubing (3 cm, long), which was then attached to Alzet pumps carrying
antibody or
88

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
vehicle (phosphate buffer saline - PBS, pH 7.4). This assembly was pre-
incubated in
PBS for 60 hrs at 37 QC to activate the pump prior to placement into the
lateral ventricle.
The assembly was surgically implanted with the use of a stereotactic apparatus
(David
Kopf Instruments) into the left lateral ventricle of each mouse at 0.4 mm
anteroposteriorly to bregma, 1.0 mm lateral to midline, and 2.5 mm
dorsoventral to the
surface of the brain under isoflurane anesthesia. Alzet osmotic pumps were
placed
under the skin by making a subcutaneous pocket with a curved, blunt ended
scissors.
Each implanted cannula was secured with dental cement along with small anchor
stainless steel screws. After the cement dried, the skin was sutured. The
antibody (2
mg/ml) or PBS in the pump was continually infused into the left lateral
ventricle of the
brain. These osmotic pumps carry a maximum of 200 pl of volume, and they pump
with
a flow rate of 3.6 I/day resulting in an infusion of 7.2 pg of antibody per
day. In each
mouse, osmotic pumps were changed once after 6 weeks of infusion. The solution

remaining in the Alzet pump was collected after its removal from each mouse
and
stored at -80 C. At the age of 9 months, all mice were sacrificed. All
surgically implanted
mice were housed singly.
[0209] Histology After 12 weeks of the treatment, P3015 mice were
anesthetized intraperitoneally with pentobarbital 200 mg/kg), followed by
perfusion with
3 U/ml heparin in cold Dulbecco's PBS. The brain was removed and cut into two
hemispheres. The left side of the brain was fixed for 24 hrs in 4%
paraformaldehyde
and transferred to 30% sucrose in PBS and stored at 4 C prior to freezing in
powdered
dry ice and stored at -80 C. Half brains were cut coronally into 50 p.m
sections with a
freezing sliding microtome and all sections were stored in 24 well plates with

cryoprotectant solution (0.2M phosphate buffered saline, 30% sucrose, 30%
ethylene
glycol) at -20 C until use. The hippocampus and cortex were dissected from the
freshly
perfused right hemisphere of each brain for biochemical analysis. All the
dissected
tissues were stored at -80 C until analyzed. The placement of the cannula into
the left
lateral ventricle was verified by mounting brain sections 300 p.m apart and
stained by
cresyl violet as previously described (Holtzman et al., 1996; Ann Neurol 39,
114-122).
89

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
The stained tissues were scanned using a NanoZoomer digital pathology system
(Hamamatsu Photonics).
[0210] Cell culture / Seeding Assay: P301S Brain Lysates and
Antibody
Treatment HEK293 cells were cultured in Dulbecco's Modified Eagle Medium
(DMEM)
supplemented with 10% fetal bovine serum, 100 pg/mL penicillin and 100 pg/mL
streptomycin. Cultures were maintained in a humidified atmosphere of 5% CO2 at
37
C. For transient transfections, HEK293 cells were plated at 250,000 cells/well
in a 12-
well plate in optimem medium and transfected using Lipofectamine 2000 reagent
and
600 ng of appropriate DNA constructs (lnvitrogen) according to manufacturer's
recommendations. Co-transfected cells received a combination of 150 ng of
RD(AK280)-CFP constructs and 450 ng of RD(AK280)-YFP constructs. 15 h later,
cells
were harvested with 0.05% trypsin for 3 minutes at 372C and then re-plated in
a 96-well
plate in quadruplicate for 15 hrs. Then, P301S brain lysates [prepared in 1X
TBS with
protease (Roche) and phosphatase inhibitors (Roche)] that were pre-incubated
with all
anti-tau monoclonal antibodies (HJ8.5, 9.3 and 9.4) or HJ3.4 antibody
(monoclonal anti-
A antibody) were added at various concentrations (0.125 pg/ml, 0.25 pg/ml, 0.5
pg/ml,
1 pg/ml and 2 gimp for 16 hrs at 4 C with rotation. To determine the seeding
activity in
the P301S mice treated for 3 months with different antibodies, RAB soluble
fractions of
all treated mice were also added to cells at various concentrations. Cells
were cultured
an additional 24 h before fixation with 4% paraformaldehyde, and FRET analysis
was
performed.
[0211] lmmunoprecipitation RAB soluble fractions from PBS or
antibody-
treated mice were incubated in the presence of mouse monoclonal anti-tau
antibodies
HJ9.3 and HJ8.5 cross-linked to protein-G-agarose beads (per kit
recommendation-
Pierce Crosslink lmmunoprecipitation kit) at 4 C with end-over-end rotation
for 24 hours.
In addition, RAB soluble fractions from antibody treated mice were incubated
in the
presence of un-conjugated protein-G-agarose beads at 42C with end-over-end
rotation
for 24 hours. 18 h later, 500 I of binding/wash buffer (Pierce) was added to
samples
and centrifuged at 2000 x g for 3 minutes. Supernatant was discarded, and this
wash
step was repeated three times. Proteins bound to beads were then eluted using
a low

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
pH elution buffer (25 I) with incubation at room temperature for 5 minutes.
Samples
were then centrifuged at 2000 x g for three minutes and supernatant collected.
This
elution step was repeated once for a total of 50 I eluates. Tau-
immunoprecipitates (IF)
containing tau aggregates were reapplied to co-transfected RD(AK)-CFP/YFP
cells at
equivalent amounts to initial brain lysates experiments for further analysis
with the
seeding assay.
[0212] Brain tissue extraction The cortex of each brain was
homogenized
in 30 I/mg of RAB buffer [100 mM MES, 1 mM EDTA, 0.5 mM Mg504, 750 mM NaCI,
20 mM NaF, 1 mM Na3VO4, supplemented by protease inhibitor (Roche) and
phosphatase inhibitor (Roche)]. In brief, the samples were centrifuged at
50,000 g for 20
min at 4 C using an Optima MAX-TL Ultracentrifuge (Beckman). The supernatants
were
collected as RAB soluble fractions and pellets were resuspended in RIPA buffer
[150
mM NaCI, 50 mM Tris, 0.5% deoxycholic acid, 1% Triton X-100, 0.5% SDS-25 mM
EDTA, pH 8.0, supplemented by protease inhibitor (Roche) and phosphatase
inhibitor
(Roche)], 30 I/mg and centrifuged at 50,000 g for 20 min at 4 C. The
supernatants
were collected as RIPA soluble fractions. The pellets were further resuspended
in 70%
formic acid, 10 I/mg and centrifuged at 50,000g for 20 min at 4 C. The
supernatants
were collected as 70% formic acid fractions. All fractions were stored in -80
C until
analyzed.
[0213] Electrophoresis and lmmunoblotting Gel electrophoresis was
performed under reducing conditions by 4-12% NuPAGE Bis-Tris gels (Invitrogen)

followed by transfer to PVDF membrane by using IBlot apparatus (Invitrogen).
70%
formic acid fractions were neutralized before loading and subjecting to gel
electrophoresis by diluting 1:3 with 1:1 mixture of 10N NaOH and
neutralization buffer
(lmol/L Tris base; 0.5 mol/L NaH4PO4). Pre-stained molecular weight standards
"SeeBlue" (Invitrogen) were included in each run. Membranes were blocked with
5%
milk in Tris buffered saline (TBS) containing 0.1% of Tween 20. Then,
membranes were
washed 3 times for 5 minutes each. Rabbit polyclonal tau antibodies (Abcam,
1:2000)
were used as primary antibodies for the detection tau in formic acid
fractions. Treated
mouse anti-tau antibodies collected before and after its infusion from osmotic
pumps
91

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
were also used as primary antibodies. The membranes were subsequently
incubated
with Goat anti-rabbit or Goat anti mouse secondary antibody (GE Healthcare,
1:2000).
All the membranes were developed with ECL prime substrate (GE Healthcare).
Bands
were visualized with G:Box Chemiluminescent Imager (Syngene).
[0214] To determine the immunoreactivity of anti-tau antibodies to
tau from
brain homogenates, RAB soluble fractions of 9 month old P3015 and 3 month old
P3015 mice, 3 month old wild type mice and 3 month old tau knockout mice
samples
were separated by SDS-PAGE followed by western blotting. Total protein of 1
lig from
each RAB soluble fraction was loaded onto 4-12% NuPAGE Bis-Tris gels
(Invitrogen)
under reducing conditions followed by transfer to nitrocellulose membrane by
using
113Iot apparatus (Invitrogen). The membranes were blocked with 5% milk in TBS
with
0.05% tween 20 (TBST) followed by incubation with primary antibodies (HJ8.5,
HJ9.3
and HJ9.4). HRP-conjugated donkey anti-mouse IgG (1:2000, Santa cruz) was used
as
secondary antibody and membranes were developed using Lumigen TMA6 (GE
Healthcare).
[0215] ELISA to detect free HJ8.5B and HJ8.5B bound to tau The
concentration of free HJ8.5B was determined in serum and CSF of mice 48hrs
after IP
or ICV administration. Ninety-six well ELISA plates were coated with 50 ng/ml
of
recombinant human tau at 4 C. ELISA plates were blocked with 4% BSA at 37 C
for 1
hr. Plates were then washed 5 times followed by incubating with serum and CSF
samples diluted in sample buffer (0.25% BSA in PBS, 300 nM Tris pH 7.4
supplemented with protease inhibitors) and incubated at 4 C overnight. The
next day,
plates were washed 8 times with PBS followed by the addition of streptavidin-
poly-
horseradish peroxidase-40 (1:6000, Fitzgerald), for 1.5 hr, in the dark, at
room
temperature. Plates were then washed 8 times with PBS and developed with Super

Slow ELISA TMB (Sigma) and read at 650 nm. Different concentration of HJ8.5B
was
used to create a standard curve that was run in each plate in addition to
serum and CSF
samples.
[0216] The concentration of HJ8.5B bound to tau was measured by
coating 96 well ELISA plates with 20 g/mlof HJ8.7 antibody at 4 C. ELISA
plates
92

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
were blocked with 4% BSA at 37 C for 1 hr. Plates were then washed 5 times
followed
by incubating with serum and CSF samples diluted in sample buffer and
incubated at 4
C overnight. The next day, plates were washed 8 times with PBS and plates were

incubated with streptavidin -poly-horseradish peroxidase-40 (1:6000,
Fitzgerald), for 1.5
hr, in the dark, at room temperature. Plates were then washed 8 times with PBS
and
developed with Super Slow ELISA TMB (Sigma) and read at 650 nm. Different
dilutions
of purified HJ8.5B complexed with recombinant tau were used to create a
standard
curve in each plate.
[0217] Tau sandwich ELISA assay To determine total tau levels,
ELISA
half 96 well plates (Costar) were coated with HJ8.7 antibody (20 g/ml) in
carbonate
buffer pH 9.6 and incubated at 4 C, overnight on a shaker. ELISA plates were
washed 5
times with PBS with a BioTek ELx405 plate washer and blocked with 4% BSA in
PBS
for 1 hr at 37 C. Plates were then washed 5 times followed by incubating wells
with
RAB, RIPA, or 70% FA biochemically extracted soluble brain tissue fractions
diluted in
sample buffer (0.25% BSA in PBS, 300 nM Tris pH 7.4 supplemented by protease
inhibitor) and incubated at 4 C. 70% FA fractions were neutralized by diluting
1:20 with
1M Tris pH 11 followed by diluting with sample buffer. The next day, plates
were
washed 8 times with PBS followed by the addition of the biotinylated mouse
monoclonal
anti-tau antibody BT-2 antibody (0.3 pg/ml, Pierce) in 0.5% BSA in PBS for 1.5
hr at 37
2 C. Plates were then washed 8 times in PBS followed by addition of
streptavidin-poly-
horseradish peroxidase-40 (1:4000), for 1.5 hr, in the dark, at room
temperature. Plates
were then washed 8 times with PBS, developed with Super Slow ELISA TMB (Sigma)

and absorbance read at 650 nm on BioTek Synergy 2 plate reader. Recombinant
human tau was used to create a standard in each plate. Negative control wells
included
omission of primary antibody in each plate. The longest recombinant human
(hTau40,
441aa) and mouse tau (mTau40, 432aa) isoforms produced in the laboratory of
Eva-
Maria Mandelkow were used as standards in the ELISA assays.
[0218] To determine the levels of human tau in 70% FA fractions,
ELISA
96 well plates were coated with mouse monoclonal antibody Tau5 (20 g/ml) and
mouse monoclonal anti-human tau specific biotinylated HT7 antibody (0.2 ug/ml,
93

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
Thermo Scientific) for detection. For mouse tau levels in the 70% FA fraction,
ELISA 96
well plates were coat with monoclonal anti-mouse tau specific HJ9.2 antibody
(20 g/ml)
and monoclonal biotinylated HJ8.7 was used for detection. Recombinant human
and
mouse tau were used for standards on each plate. To determine phospho tau
levels at
positions 5er202 and Thr205, ELISA half 96 well plates were coated with mouse
monoclonal HJ8.7 antibody (20 g/ml) and biotinylated AT8 antibody (0.2 ug/ml,

Thermo Scientific) was used as detection antibody.
[0219] lmmunohistochemistry To detect the presence of abnormally
phosphorylated tau in the brain, three 50 pm coronal brain sections spaced 300
pm
apart from all treated mice were assessed. The brain sections were blocked
with 3%
milk in Tris-buffered saline (TBS) and 0.25% (vol/vol) Triton-X followed by
incubation at
4 C overnight with the biotinylated AT8 antibody (Thermo Scientific, 1:500)
which
recognizes tau phosphorylated at ser202 and thr205. Biotinylated PHF1 antibody

(1:200) which recognizes abnormally phosphorylated tau at residues ser396 and
ser404
was also used to determine the correlation between AT8 and PHF1 antibody
staining.
For correlation studies, mice (N=6) were randomly selected from the HJ8.5,
HJ9.3, and
PBS-treated groups. The stained tissues were scanned using the NanoZoomer
digital
pathology system. To determine the correlation between the AT8 staining and
activated
microglial staining, brain sections from selected mice of all the treated
groups (N= 6),
were blocked with 10% normal goat serum in TBS with 0.25% (vol/vol) Triton-X
was
incubated with a rat anti-mouse CD68 antibody (AbD SeroTec, 1:500) at 4 C
overnight.
The sections were then incubated with biotinylated goat anti-rat IgG antibody,
mouse
adsorbed (Vector, 1:2000). All sections were scanned with a NanoZoomer slide
scanner
(Hamamatsu Photonics). All images were exported by using NDP viewer software
and
quantified by using ImageJ software (National Institutes of Health). For AT8
staining, 3
brain sections from each mouse separated by 300 pm, corresponding
approximately to
sections at Bregma coordinates -1.4, -1.7, and -2.0 mm in the mouse brain
atlas were
used. These sections were used to determine the percentage of area occupied by

abnormally phosphorylated biotinylated AT8 antibody staining. All converted
images
were uniformly thresholded to quantify AT8 staining and the average of all
three
94

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
sections was used to determine the percentage of area covered by abnormally
phosphorylated tau staining for each mouse. For PH F-1 and CD68 staining, two
brain
sections from each mouse were used, separated by 300 pm and correspond to
bregma
coordinates -2.3 and -2.6 mm in the mouse brain atlas. To determine ThioS
staining,
brain sections from randomly selected mice from all the treated groups (N. 6)
were
stained in ThioS in 50% ethanol (0.25 mg/ml) for 3 min, followed by washing in
50%
ethanol and distilled water. Slices were then mounted, dried and images were
assessed
by microscopy with the Nanozoomer. Two brain sections from each mouse were
used
as described adjacent to those used for PH F-1 and CD68 staining.
[0220] Semi-Denaturing-Agarose Gel Electrophoresis (SDD-AGE) For
separation of tau species present in the different RAB soluble fractions of 3
month old
tau knockout (KO), 3 months old wild type (WT), 3 months old P301S and 9 month
old
PBS-treated P301S mice, the previously described Semi-Denaturing Detergent-
Agarose
Gel Electrophoresis (SDD-AGE) method was employed with minor modifications.
Samples were run on horizontal 1.5% agarose gels in Buffer G (20 mM Tris, 200
mM
Glycine) with 0.2% SDS. Samples were incubated in the sample buffer (60 mM
Tris-HCI
pH 6.8, 0.2% SDS, 5% glycerol, and 0.05% bromphenol blue) for 7 min at RT.
After the
electrophoresis, proteins were transferred from gels to lmmobilon-P PVDF
sheets
(Millipore) at 4 C in Laemmli Buffer (Buffer G/0.1 /0 SDS). Membranes were
blotted
using an anti-tau specific rabbit polyclonal antibody (Abcam) at 1:2000. Blots
were
developed using the GE ECL Plus system.
[0221] lmmunofluorescence HEK293 cells were plated at 75,000 cells/
well in 24 well plates coated with poly D-lysine. To determine whether anti-
tau
antibodies used can detect tau species taken up by the HEK293 cells, the cells
were
treated with P301S brain lysates for 2 hrs, followed by washing 3x with PBS,
fixed with
4% paraformaldehyde for 15 min at room temperature followed by washing 3 times
with
PBS at room temperature. Cells were permeabilized with 0.1% Triton X-100 for
10 min,
washed 3 times with PBS, then blocked with 0.25% Triton X-100 in PBS
containing 10%
normal goat serum and 20 mg/ml BSA. Then cells were incubated with anti-mouse
secondary antibody conjugated with Alexa-fluor 546. To determine whether
antibody

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
can enter the cells, P3015 brain lysates were pre-incubated with and without
the
different anti-tau antibodies HJ8.5, HJ9.3, and HJ9.4 or the HJ3.4 antibody to
A. The
lysates were then added to HEK293 cells for 2 hrs, fixed and permeabilized.
Secondary
antibody conjugated with Alexa-fluor 546 was used to identify the antibodies.
4',6'-
diamidino-2-phenylindole (DAPI; shown in blue) was used for nuclear stain. All
the
images were captured by using a Zeiss LSM5 confocal microscope (Zeiss).
[0222] Statistical analysis of pathological and biochemical data
All data are
presented as mean SEM, and different conditions were compared using one-way
ANOVA followed by Dunnett's post hoc test to compare controls with treatment
groups.
Statistical significance was set at P <0.05. Statistics were performed using
GraphPad
Prism 5.04 for Windows (Graph Pad Software Inc.). For quantitative assessment
of AT8
staining, gender is a significant factor so results were adjusted by gender
using SAS
version 9.2 software.
[0223] Statistical analysis applying treatment and gender as
factors The
control group (PBS and HJ3.4) mean was compared with each treatment group.
(mean
of PBS+ mean of HJ3.4)/2 VS mean of treatment). Two-way ANOVA was used to test

whether gender and treatment are significant factors, which is achieved by
PROC GLM
in SAS Version 9.2 and their p Values are shown in Table 7 . A contrast
statement was
used in PROC GLM of SAS Version 9.2 to access all comparisons. Gender as an
adjustment factor in the two-way ANOVA was applied and p Values before/after
the
adjustment are shown in FIG. 380.
[0224] Behavioral tests Mice were assessed on locomotor activity
and
exploratory behaviors and on a battery of sensorimotor measures and the
rotarod to
provide additional control data for interpreting the results of the
conditional fear test,
which was used to evaluate cognitive function. The conditioned fear test was
conducted
last in the series of tests to preclude effects of brief footshocks on other
behavioral
indices.
[0225] Holeboard exploration, sensorimotor battery and rotarod. All
mice
were evaluated on the holeboard exploration test where total ambulations
(whole body
movements) and hole pokes were quantified over a 30-min period and provided
indices
96

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
of locomotor activity and exploration. The protocol involved the use of a
computerized
holeboard apparatus (41 x 41 x 38.5 cm high) containing 4 corner and 4 side
holes, with
a side hole being equidistant between the corner holes (Learning Holeboard;
MotorMonitor, Kinder Scientific, LLC, Poway, CA). Photobeam instrumentation
was
used to quantify total ambulations and exploratory hole pokes during the test
session.
This procedure has served as the habituation component of our general
holeboard
exploration/olfactory preference test. The mice were also tested on a battery
of seven
sensorimotor measures that were used to assess balance (ledge, platform), co-
ordination (pole, 60 and 90 inclined screens), strength (inverted screen),
and initiation
of movement out of a small circumscribed area (walking initiation). This
battery was
used in previous publications and greater procedural details may be found in
(Wozniak
et al. (2004; Neurobiol Dis 17, 403-414). The rotarod test was similar to
previously-
published methods and included three types of trials: 1) stationary rod (60 s
maximum;
2) constant speed rotarod (2.5 rpm for 60 s maximum; and 3) accelerating
rotarod (2.5-
10.5 rpm over 0-180 s). Our protocol consisted of testing each mouse on one
stationary
rod trial, two constant speed rotarod trials, and two accelerating rotarod
trials for each of
three test sessions that were separated by 3 days to limit motor learning.
[0226] Conditioned fear. Mice were evaluated on the conditioned
fear test,
which was the last behavioral measure conducted. Briefly, the mice were
trained and
tested in two Plexiglas conditioning chambers (26 cm x 18 cm, and 18 cm high)
(Med-
Associates, St. Albans, VT) with each chamber containing distinct and
different visual,
odor, and tactile cues. Each mouse was placed into the conditioning chamber
for a 5-
min trial and freezing behavior was quantified during a 2-min baseline period.
Beginning
at 3 min and at 60-s intervals thereafter, the mice were exposed to 3 tone-
shock
pairings where each pairing included a 20-s presentation of an 80 dB tone
(conditioned
stimulus; CS) consisting of broadband white noise followed by a 1.0 mA
continuous
footshock (unconditioned stimulus; CS) presented during the last second of the
tone.
Broadband white noise was used instead of a frequency-specific tone in an
effort to
avoid possible auditory deficits that might occur with age. The mice were
placed back
into the conditioning chamber the following day and freezing behavior was
quantified
97

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
over an 8-min period to evaluate contextual fear conditioning. Twenty four
hours later,
the mice were placed into the other chamber containing different cues and
freezing
behavior was quantified during a 2-min "altered context" baseline and over the

subsequent 8 min, during which time the auditory cue (tone; CS) was presented.

Freezing was quantified using FreezeFrame image analysis software
(Actimetrics,
Evanston, IL), which allowed for simultaneous visualization of behavior while
adjusting a
"freezing threshold," which categorized behavior as freezing or not freezing
during 0.75
s intervals. Freezing was defined as no movement except for that associated
with
normal respiration, and the data were presented as percent of time spent
freezing. To
assess the extent of contextual fear conditioning, we conducted analyses
within each
treatment group which involved comparing the percent time spent freezing
averaged
over the 2-min baseline on day 1 with the averaged percent time spent freezing
during
the first 2 min of the contextual fear test on day 2, as well as with freezing
levels
averaged across the entire 8-min session. Shock sensitivity was evaluated
following
completion of the conditioned fear testing, according to previously described
procedures
in Khuchua et al. (2003; Neuroscience 119, 101-111).
[0227] Statistical Analyses of behavioral data Analysis of variance
(ANOVA) models were typically used to analyze the behavioral data (Systat 12,
Systat
Software, Chicago, IL). The conditioned fear data were analyzed using repeated

measures (rm) ANOVA models containing one between-subjects variable
(Treatment)
and one within-subjects (repeated measures) variable (Minutes). The Huynh-
Feldt
adjustment of alpha levels was utilized for all within-subjects effects
containing more
than two levels to protect against violations of sphericity/compound symmetry
assumptions underlying rmANOVA models. Planned comparisons between the
PBS+HJ3.4 control group and each of the three other antibody treatment groups
(i.e.,
HJ8.5, HJ9.3, HJ9.4) were conducted within ANOVA models for testing certain
key
hypotheses. In other instances, pair-wise comparisons were conducted following

relevant, significant overall ANOVA effects, which were subjected to
Bonferroni
correction when appropriate. Pearson's correlation coefficient (r) was also
calculated
98

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
between the total ambulations recorded during the holeboard test and the
percent time
spent freezing during the contextual fear test on day 2.
Example 16. Tau ELISA Assay
[0228] An ELISA assay was developed in order to detect the presence
of
pathological tau aggregates in plasma samples of patients. Antibodies used in
this
assay include mouse monoclonal anti-tau HJ9.3 and HJ9.2. HJ9.3 is biotinylated
using
One-step Antibody Biotinylation Kit (HJ9.3-Bio). This sandwich ELISA utilizes
HJ9.3 and
HJ9.2, at equivalent concentration, as capture antibodies. 96-well half area
plates
(Costar 3690) are coated with 20 pg/m1 of HJ9.2/HJ9.3 prepared in bicarbonate
buffer
pH 9.6 (50 I/well) and incubated at 4 C overnight. Following a blocking step
using 4 %
BSA/PBS, plasma samples (diluted 1:4 in sample buffer: 0.25% BSA/PBS, 300nM
Tris
PH 7.4-8.0, lx protease inhibitors) are applied in triplicates to wells (50
1/well). Plates
are then incubated at 4 C overnight. For detection, HJ9.3-Bio prepared in
0.5%
BSA/PBS at 0.3 g/m1 was added to wells for 1.5 hr at 37 C. A secondary
streptavidin-
polyHRP40 antibody at 1:4,000 dilution in 0.5% BSA/PBS (50 I/well and 1.5 hr
in dark
on a shaker at RT) is used for final detection through an enzymatic reaction
using TMB
super slow substrate. The ELISA has been designed to optimize detection of
rare
species in plasma. Initial embodiments included coating the surface of the
ELISA plate
with antibody pairs to optimize trapping of aggregates. However it would be
equally
plausible to use antibody coated beads from larger volumes of fluid samples to
increase
the sensitivity of the assay. Negative plasma collected from healthy young
participants
was used to calculate the background signal of the assay. Tau seeds presence
in the
experimental samples is reported as fold induction over signal from negative
plasma.
[0229] A set of plasma samples from pre-clinical and Alzheimer's
Disease
(AD) patients previously tested with the disclosed seeding assay were used to
validate
the sandwich tau ELISA assay. 12 control patients (CDR 0) with no seeding
activity
(Negative) and 12 patients (CDR > 0) with seeding activity (Positive) were
tested using
the newly developed ELISA assay. These patients were previously determined to
have
seeding activity or not in CSF and plasma based on a biosensor cellular assay.
In this
99

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
cellular assay, RD fragments of the tau protein containing the AK280 mutation
are fused
to cyan or yellow fluorescent protein. This enables detection of aggregation
by
measuring fluorescence resonance energy transfer via FRET. Extracellular
aggregates
are brought into the cell and trigger intracellular aggregation of the tau
FRET reporter
proteins.
[0230] As shown in FIG. 45, no tau aggregates were detected in the
plasma of patients with negative seeding activity compared to the clear tau
presence of
seeds in the plasma of AD patients with positive seeding activity. This cell-
free based
assay could be used in a more clinical setting as a non-invasive diagnostic
tool for many
tauopathies including Alzheimer's Disease. Further, it could allow detection
of those
with incipient pathology who are destined to develop dementia, facilitating
clinical trial
design by enriching a sample population. Finally, it could be used to monitor
efficacy of
anti-tau or other anti-dementia therapies.
Example 17
[0231] A cellular propagation assay was set up to measure the
propagation of tau aggregates from one population to another. A fragment of
tau
comprised of the repeat domain (RD) was used either as an untagged form with
two
disease-associated mutations (LM: P301L/V337M) to promote aggregation of the
CFP-
tagged form, or one disease-associated mutation (AK: AK280). One group of
cells was
transfected with RD(LM) and RD(AK280)-CFP, and another was transfected with
RD(AK280)-YFP. FRET was recorded on a fluorescence plate reader from cells
grown
in quadruplicate in a 96-well format. FRET signal derives from RD-CFP
aggregates
transferring to cells containing RD-YFP, and vice-versa. Multiple antibodies
were added
to the medium at various dilutions indicated. The starting concentration of
antibody was
-1mg/ml. For example, a 10-3dilution indicates a final concentration of -1
g/ml. After
24h the cells were fixed and FRET measurements recorded. Data for individual
antibodies are presented in FIG. 47. Some antibodies were very potent at
preventing
trans-cellular propagation of aggregation (e.g. HJ8.2, HJ9.1). Others were
effective in a
more intermediate fashion (e.g. HJ9.3), and some were essentially not
effective (HJ8.7).
100

CA 02877397 2014-12-18
WO 2014/008404 PCT/US2013/049333
In each graph, the first bar represents medium without added antibody,
representing
baseline efficiency of propagation.
[0232] To test for synergy of antibodies, effects on propagation
were
determined in the setting of individual antibodies diluted over an indicated
concentration
range, or antibodies were mixed at an equimolar ratio and then titrated over
the same
range. Some pairs were strongly synergistic (e.g. HJ9.3/9.4), while others
interfered
with one another (HJ8.5/9.1) (FIG. 48).
[0233] The effect of an antibody on tau aggregate uptake may also
be
measured by flow cytometry. Cells were exposed to recombinant RD fibrils that
were
chemically labeled with a fluorescent dye. After trypsinization and
dispersion, the cells
were counted using a flow cytometer. HJ9.3 dose-dependently reduces the number
of
fluorescently labeled cells, indicating inhibition of aggregate uptake (FIG.
50).
101

Representative Drawing

Sorry, the representative drawing for patent document number 2877397 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2013-07-03
(87) PCT Publication Date 2014-01-09
(85) National Entry 2014-12-18
Examination Requested 2018-06-28
Dead Application 2022-10-21

Abandonment History

Abandonment Date Reason Reinstatement Date
2019-10-24 R30(2) - Failure to Respond 2020-10-23
2021-10-21 FAILURE TO PAY FINAL FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2014-12-18
Maintenance Fee - Application - New Act 2 2015-07-03 $100.00 2014-12-18
Maintenance Fee - Application - New Act 3 2016-07-04 $100.00 2016-06-17
Maintenance Fee - Application - New Act 4 2017-07-04 $100.00 2017-06-16
Maintenance Fee - Application - New Act 5 2018-07-03 $200.00 2018-06-14
Request for Examination $800.00 2018-06-28
Maintenance Fee - Application - New Act 6 2019-07-03 $200.00 2019-06-18
Maintenance Fee - Application - New Act 7 2020-07-03 $200.00 2019-11-19
Reinstatement - failure to respond to examiners report 2020-10-23 $200.00 2020-10-23
Maintenance Fee - Application - New Act 8 2021-07-05 $204.00 2021-06-16
Maintenance Fee - Application - New Act 9 2022-07-04 $203.59 2022-06-21
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
WASHINGTON UNIVERSITY
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Maintenance Fee Payment 2019-11-19 1 33
Office Letter 2019-11-20 1 22
Office Letter 2019-11-20 1 23
Change of Agent 2019-12-19 3 80
Office Letter 2020-01-08 1 184
Office Letter 2020-01-08 1 193
Reinstatement / Amendment 2020-10-23 14 610
Description 2020-10-23 101 5,287
Abstract 2014-12-18 1 50
Claims 2014-12-18 3 87
Drawings 2014-12-18 98 7,995
Description 2014-12-18 101 5,093
Cover Page 2015-02-11 1 23
Request for Examination 2018-06-28 2 44
Amendment 2018-06-28 4 84
Description 2015-05-19 101 5,323
Claims 2018-06-28 2 39
Examiner Requisition 2019-04-24 3 150
Change of Agent 2019-10-23 1 31
PCT 2014-12-18 4 165
Assignment 2014-12-18 4 106
Correspondence 2015-01-30 1 34
Prosecution-Amendment 2015-05-19 6 245

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :